321 results on '"Vittorio, Rosti"'
Search Results
2. VEGFA rs3025039 is associated with phenotype severity of myelofibrosis‐type megakaryocyte dysplasia
- Author
-
Giovanni Barosi, Paolo Catarsi, Rita Campanelli, Margherita Massa, Adriana Carolei, Carlotta Abbà, Annalisa deSilvestri, Robert Peter Gale, and Vittorio Rosti
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
3. NATURAL HISTORY OF RALD: A 20 YEAR FOLLOW-UP OF A NRAS MUTATED PATIENT EXCLUDING A MALIGNANT PROGRESSION
- Author
-
Enrico Attardi, Beatrice Rivalta, Cristina Cifaldi, Vittorio Rosti, Lucia Pacillo, Hajro Hajrullaj, Silvia Di Cesare, Matteo Luciani, Federica Barzaghi, Andrea Finocchi, Gigliola Di Matteo, Alessandro Aiuti, Franco Locatelli, Maria Teresa Voso, Giuseppe Palumbo, and Caterina Cancrini
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
4. Circulating Mesenchymal Stromal Cells in Patients with Infantile Hemangioma: Evaluation of Their Functional Capacity and Gene Expression Profile
- Author
-
Carlotta Abbà, Stefania Croce, Chiara Valsecchi, Elisa Lenta, Rita Campanelli, Alessia C. Codazzi, Valeria Brazzelli, Adriana Carolei, Paolo Catarsi, Gloria Acquafredda, Antonia Apicella, Laura Caliogna, Micaela Berni, Savina Mannarino, Maria A. Avanzini, Vittorio Rosti, and Margherita Massa
- Subjects
infantile hemangioma ,mesenchymal stromal cells ,propranolol ,angiogenesis ,Cytology ,QH573-671 - Abstract
We previously published that in patients with infantile hemangioma (IH) at the onset (T0) colony forming unit-fibroblasts (CFU-Fs) are present in in vitro cultures from PB. Herein, we characterize these CFU-Fs and investigate their potential role in IH pathogenesis, before and after propranolol therapy. The CFU-F phenotype (by flow cytometry), their differentiation capacity and ability to support angiogenesis (by in vitro cultures) and their gene expression (by RT-PCR) were evaluated. We found that CFU-Fs are actual circulating MSCs (cMSCs). In patients at T0, cMSCs had reduced adipogenic potential, supported the formation of tube-like structures in vitro and showed either inflammatory (IL1β and ESM1) or angiogenic (F3) gene expression higher than that of cMSCs from CTRLs. In patients receiving one-year propranolol therapy, the cMSC differentiation in adipocytes improved, while their support in in vitro tube-like formation was lost; no difference was found between patient and CTRL cMSC gene expressions. In conclusion, in patients with IH at T0 the cMSC reduced adipogenic potential, their support in angiogenic activity and the inflammatory/angiogenic gene expression may fuel the tumor growth. One-year propranolol therapy modifies this picture, suggesting cMSCs as one of the drug targets.
- Published
- 2024
- Full Text
- View/download PDF
5. Elevated plasma EDA fibronectin in primary myelofibrosis is determined by high allele burden of JAK2V617F mutation and strongly predicts splenomegaly progression
- Author
-
Alessandro Malara, Cristian Gruppi, Margherita Massa, Maria Enrica Tira, Vittorio Rosti, Alessandra Balduini, and Giovanni Barosi
- Subjects
primary myelofibrosis ,splenomegaly ,neoangiogenesis ,extra domain A ,fibronectin ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
In primary myelofibrosis, extra-domain A fibronectin (EDA-FN), the result of alternative splicing of FN gene, sustains megakaryocyte proliferation and confers a pro-inflammatory phenotype to bone marrow cell niches. In this work we assessed the levels of circulating EDA-FN in plasma samples of 122 patients with primary myelofibrosis. Patients with a homozygous JAK2V617F genotype displayed the higher level of plasma EDA-FN. Increased EDA-FN levels were associated with anemia, elevated high-sensitivity C-reactive protein, bone marrow fibrosis and splanchnic vein thrombosis at diagnosis. While no correlation was observed with CD34+ hematopoietic stem cell mobilization, elevated blood level of EDA-FN at diagnosis was a predictor of large splenomegaly (over 10 cm from the left costal margin) outcome. Thus, EDA-FN expression in primary myelofibrosis may represent the first marker of disease progression, and a novel target to treat splenomegaly.
- Published
- 2022
- Full Text
- View/download PDF
6. Long-term safety and efficacy of givinostat in polycythemia vera: 4-year mean follow up of three phase 1/2 studies and a compassionate use program
- Author
-
Alessandro Rambaldi, Alessandra Iurlo, Alessandro M. Vannucchi, Bruno Martino, Attilio Guarini, Marco Ruggeri, Nikolas von Bubnoff, Marianna De Muro, Mary Frances McMullin, Stefania Luciani, Vincenzo Martinelli, Axel Nogai, Vittorio Rosti, Alessandra Ricco, Paolo Bettica, Sara Manzoni, and Silvia Di Tollo
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Polycythemia vera (PV) is a BCR-ABL1-negative myeloproliferative neoplasm (MPN) characterized by excessive proliferation of erythroid, myeloid, and megakaryocytic components in the bone marrow, mainly due to a Janus kinase 2 gene mutation (JAK2 V617F). Givinostat, a histone-deacetylase inhibitor that selectively targets JAK2 V617F cell growth, has demonstrated good efficacy and safety in three phase 1/2 studies in patients with PV. This manuscript focuses on the 4-year mean (2.8 year median) follow-up of an open-label, long-term study that enrolled 51 patients with PV (out of a total of 54 with MPN) who received clinical benefit from givinostat in these previous studies or on compassionate use, and who continued to receive givinostat at the last effective and tolerated dose. The primary objectives are to determine givinostat’s long-term safety and tolerability, and efficacy evaluated by the investigators according to internationally recognized response criteria. During follow-up, only 10% of PV patients reported Grade 3 treatment-related adverse events (AEs), while none had Grade 4 or 5 treatment-related AEs. The overall response rate for the duration of follow-up was always greater than 80% in patients with PV. In conclusion, givinostat demonstrated a good safety and efficacy profile in patients with PV, data supporting long-term use in this population.
- Published
- 2021
- Full Text
- View/download PDF
7. VEGFA rs3025020 Polymorphism Contributes to CALR-Mutation Susceptibility and Is Associated with Low Risk of Deep Vein Thrombosis in Primary Myelofibrosis
- Author
-
Laura Villani, Vittorio Rosti, Margherita Massa, Rita Campanelli, Paolo Catarsi, Adriana Carolei, Carlotta Abbà, Annalisa de Silvstri, Robert Peter Gale, and Giovanni Barosi
- Subjects
primary myelofibrosis ,vegfa polymorphism ,rs3025020 ,deep vein thrombosis ,calr mutation ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background Single nucleotide polymorphisms (SNPs) in vascular endothelial growth factor A (VEGFA) are associated with susceptibility to several diseases including cancer. Correlations between VEGFA rs3025020 genotypes with clinical and laboratory features of primary myelofibrosis (PMF) are unstudied. Methods DNA was analyzed by real-time polymerase chain reaction for VEGFA rs3025020 genotypes in a cohort of 844 subjects with PMF and in two cohorts of normal subjects (N = 247 and N = 107). Results Frequency of rs3025020 minor allele (T) was not significantly different in subjects with PMF compared with normals; however, the T-allele was more frequent in PMF subjects with a calreticulin (CALR)-mutated genotype compared with normals (35 vs. 27%; OR = 1.47 [95% CI, 1.09, 1.98] p = 0.011), especially in subjects with a CALR-type 2/type 2-like mutation (43 vs. 27%; OR = 2.01 [1.25, 3.24] p = 0.004). CALR mutants with the rs3025020 TT genotype had higher CXCR4 expression on CD34-positive blood cells, and those who carried CT/TT genotypes had lower platelet concentrations compared with other genotypes at diagnosis. Overall, subjects with the rs3025020 CT/TT genotype had a lower cumulative incidence of deep vein thrombosis in typical sites (1.6 vs. 4.2%; OR = 0.37 [0.15, 0.90] p = 0.029) and longer interval from diagnosis to first thrombosis (HR = 0.37 [0.14, 0.95] p = 0.039). Conclusion Persons with PMF and the VEGFA rs3025020 minor T-allele are more likely to have a CALR mutation compared with other somatic driver mutations and lower cumulative incidence and hazard for deep vein thrombosis in typical sites.
- Published
- 2021
- Full Text
- View/download PDF
8. Bi-allelic variants in the ESAM tight-junction gene cause a neurodevelopmental disorder associated with fetal intracranial hemorrhage
- Author
-
Mauro Lecca, Davut Pehlivan, Damià Heine Suñer, Karin Weiss, Thibault Coste, Markus Zweier, Yavuz Oktay, Nada Danial-Farran, Vittorio Rosti, Maria Paola Bonasoni, Alessandro Malara, Gianluca Contrò, Roberta Zuntini, Marzia Pollazzon, Rosario Pascarella, Alberto Neri, Carlo Fusco, Dana Marafi, Tadahiro Mitani, Jennifer Ellen Posey, Sadik Etka Bayramoglu, Alper Gezdirici, Jessica Hernandez-Rodriguez, Emilia Amengual Cladera, Elena Miravet, Jorge Roldan-Busto, María Angeles Ruiz, Cristofol Vives Bauzá, Liat Ben-Sira, Sabine Sigaudy, Anaïs Begemann, Sheila Unger, Serdal Güngör, Semra Hiz, Ece Sonmezler, Yoav Zehavi, Michael Jerdev, Alessandra Balduini, Orsetta Zuffardi, Rita Horvath, Hanns Lochmüller, Anita Rauch, Livia Garavelli, Elisabeth Tournier-Lasserve, Ronen Spiegel, James R. Lupski, and Edoardo Errichiello
- Subjects
Genetics ,Genetics (clinical) - Abstract
The blood-brain barrier (BBB) is an essential gatekeeper for the central nervous system and incidence of neurodevelopmental disorders (NDDs) is higher in infants with a history of intracerebral hemorrhage (ICH). We discovered a rare disease trait in thirteen individuals, including four fetuses, from eight unrelated families associated with homozygous loss-of-function variant alleles of ESAM which encodes an endothelial cell adhesion molecule. The c.115del (p.Arg39Glyfs∗33) variant, identified in six individuals from four independent families of Southeastern Anatolia, severely impaired the in vitro tubulogenic process of endothelial colony-forming cells, recapitulating previous evidence in null mice, and caused lack of ESAM expression in the capillary endothelial cells of damaged brain. Affected individuals with bi-allelic ESAM variants showed profound global developmental delay/unspecified intellectual disability, epilepsy, absent or severely delayed speech, varying degrees of spasticity, ventriculomegaly, and ICH/cerebral calcifications, the latter being also observed in the fetuses. Phenotypic traits observed in individuals with bi-allelic ESAM variants overlap very closely with other known conditions characterized by endothelial dysfunction due to mutation of genes encoding tight junction molecules. Our findings emphasize the role of brain endothelial dysfunction in NDDs and contribute to the expansion of an emerging group of diseases that we propose to rename as “tightjunctionopathies.”
- Published
- 2023
- Full Text
- View/download PDF
9. Shared and Distinctive Ultrastructural Abnormalities Expressed by Megakaryocytes in Bone Marrow and Spleen From Patients With Myelofibrosis
- Author
-
Maria Zingariello, Vittorio Rosti, Alessandro M. Vannucchi, Paola Guglielmelli, Maria Mazzarini, Giovanni Barosi, Maria Luisa Genova, and Anna Rita Migliaccio
- Subjects
myeloproliferative neoplasms ,myelofibrosis ,megakaryocytes ,ultrastructural analyses ,cell metabolism ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Numerous studies have documented ultrastructural abnormalities in malignant megakaryocytes from bone marrow (BM) of myelofibrosis patients but the morphology of these cells in spleen, an important extramedullary site in this disease, was not investigated as yet. By transmission-electron microscopy, we compared the ultrastructural features of megakaryocytes from BM and spleen of myelofibrosis patients and healthy controls. The number of megakaryocytes was markedly increased in both BM and spleen. However, while most of BM megakaryocytes are immature, those from spleen appear mature with well-developed demarcation membrane systems (DMS) and platelet territories and are surrounded by platelets. In BM megakaryocytes, paucity of DMS is associated with plasma (thick with protrusions) and nuclear (dilated with large pores) membrane abnormalities and presence of numerous glycosomes, suggesting a skewed metabolism toward insoluble polyglucosan accumulation. By contrast, the membranes of the megakaryocytes from the spleen were normal but these cells show mitochondria with reduced crests, suggesting deficient aerobic energy-metabolism. These distinctive morphological features suggest that malignant megakaryocytes from BM and spleen express distinctive metabolic impairments that may play different roles in the pathogenesis of myelofibrosis.
- Published
- 2020
- Full Text
- View/download PDF
10. Role of TGF‐β1/miR‐382‐5p/SOD2 axis in the induction of oxidative stress in CD34+ cells from primary myelofibrosis
- Author
-
Chiara Rossi, Roberta Zini, Sebastiano Rontauroli, Samantha Ruberti, Zelia Prudente, Greta Barbieri, Elisa Bianchi, Simona Salati, Elena Genovese, Niccolò Bartalucci, Paola Guglielmelli, Enrico Tagliafico, Vittorio Rosti, Giovanni Barosi, Alessandro M. Vannucchi, Rossella Manfredini, and the AGIMM (AIRC‐Gruppo Italiano Malattie Mieloproliferative) investigators
- Subjects
miR‐382‐5p ,oxidative stress ,primary myelofibrosis ,reactive oxygen species ,superoxide dismutase ,TGF‐β1 signaling ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by an excessive production of pro‐inflammatory cytokines resulting in chronic inflammation and genomic instability. Besides the driver mutations in JAK2, MPL, and CALR genes, the deregulation of miRNA expression may also contribute to the pathogenesis of PMF. To this end, we recently reported the upregulation of miR‐382‐5p in PMF CD34+ cells. In order to unveil the mechanistic details of the role of miR‐382‐5p in pathogenesis of PMF, we performed gene expression profiling of CD34+ cells overexpressing miR‐382‐5p. Among the downregulated genes, we identified superoxide dismutase 2 (SOD2), which is a predicted target of miR‐382‐5p. Subsequently, we confirmed miR‐382‐5p/SOD2 interaction by luciferase assay and we showed that miR‐382‐5p overexpression in CD34+ cells causes the decrease in SOD2 activity leading to reactive oxygen species (ROS) accumulation and oxidative DNA damage. In addition, our data indicate that inhibition of miR‐382‐5p in PMF CD34+ cells restores SOD2 function, induces ROS disposal, and reduces DNA oxidation. Since the pro‐inflammatory cytokine transforming growth factor‐β1 (TGF‐β1) is a key player in PMF pathogenesis, we further investigated the effect of TGF‐β1 on ROS and miR‐382‐5p levels. Our data showed that TGF‐β1 treatment enhances miR‐382‐5p expression and reduces SOD2 activity leading to ROS accumulation. Finally, inhibition of TGF‐β1 signaling in PMF CD34+ cells by galunisertib significantly reduced miR‐382‐5p expression and ROS accumulation and restored SOD2 activity. As a whole, this study reports that TGF‐β1/miR‐382‐5p/SOD2 axis deregulation in PMF cells is linked to ROS overproduction that may contribute to enhanced oxidative stress and inflammation. Our results suggest that galunisertib may represent an effective drug reducing abnormal oxidative stress induced by TGF‐β1 in PMF patients. Database linking GEO: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE103464.
- Published
- 2018
- Full Text
- View/download PDF
11. Myelofibrosis-type megakaryocyte dysplasia (MTMD) as a distinct category of BCR::ABL-negative myeloproliferative neoplasms. Challenges and perspectives
- Author
-
Giovanni Barosi, Vittorio Rosti, and Robert Peter Gale
- Subjects
Cancer Research ,Oncology ,Hematology - Abstract
In this Perspective, we discuss criteria for defining a new disease entity or variant of a recognized disease or disorder. We do so in the context of the current topography of the BCR::ABL-negative myeloproliferative neoplasms (MPNs) where two new variants are reported: clonal megakaryocyte dysplasia with normal blood values (CMD-NBV) and clonal megakaryocyte dysplasia with isolated thrombocytosis (CMD-IT). The cardinal feature of these variants is bone marrow megakaryocyte hyperplasia and atypia corresponding the WHO histological criteria for primary myelofibrosis (myelofibrosis-type megakaryocyte dysplasia-MTMD). Persons with these new variants have a different disease course and features from others in the MPN domain. In a broader context we suggest myelofibrosis-type megakaryocyte dysplasia defines a spectrum of related MPN variants including CMD-NBV, CMD-IT, pre-fibrotic myelofibrosis and overt myelofibrosis, which differ from polycythemia vera and essential thrombocythemia. Our proposal needs external validation and we stress the need for a consensus definition of the megakaryocyte dysplasia which is the hallmark of these disorders.
- Published
- 2023
- Full Text
- View/download PDF
12. Supporting data on in vitro cardioprotective and proliferative paracrine effects by the human amniotic fluid stem cell secretome
- Author
-
Carolina Balbi, Kirsten Lodder, Ambra Costa, Silvia Moimas, Francesco Moccia, Tessa van Herwaarden, Vittorio Rosti, Francesca Campagnoli, Agnese Palmeri, Pierangela De Biasio, Francesco Santini, Mauro Giacca, Marie-Josè Goumans, Lucio Barile, Anke M. Smits, and Sveva Bollini
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 ,Science (General) ,Q1-390 - Abstract
The data and information presented here refer to the research article entitled: “Reactivating endogenous mechanisms of cardiac regeneration via paracrine boosting with the human amniotic fluid stem cell secretome” (Balbi et al., 2019, Apr 04). This dataset illustrates the in vitro paracrine effect exerted by the human amniotic fluid stem cell secretome on rodent neonatal cardiomyocytes, human endothelial progenitors and different subsets of cardiac progenitor cells. Cytokine/chemokine profiling of the human amniotic fluid stem cell secretome is provided as well. This data can provide useful insights in regenerative medicine as demonstrating the in vitro cardioprotective and proliferative secretory paracrine potential of human fetal stem cells. Keywords: Paracrine effect, Human amniotic fluid stem cells, Cardiac progenitor cells, Neonatal cardiomyocytes, Endothelial progenitors, Proliferation, Cardioprotection, Angiogenesis
- Published
- 2019
- Full Text
- View/download PDF
13. Constitutive STAT5 phosphorylation in CD34+ cells of patients with primary myelofibrosis: Correlation with driver mutation status and disease severity.
- Author
-
Carlotta Abbà, Rita Campanelli, Paolo Catarsi, Laura Villani, Vittorio Abbonante, Melania Antonietta Sesta, Giovanni Barosi, Vittorio Rosti, and Margherita Massa
- Subjects
Medicine ,Science - Abstract
Primary Myelofibrosis (PMF) is a myeloproliferative disorder associated with JAK2V617F, Calreticulin (CALR) indels, and MPLW515L/K mutations activating the tyrosine kinase JAK2 and its downstream signaling pathway. The nature of signaling abnormalities in primary cells from PMF patients is poorly understood, since most of the work has been performed in cell lines or animal models. By flow cytometry we measured constitutive and cytokine induced phosphorylation of STAT5, STAT3, and ERK1/2 in circulating CD34+ cells from 57 patients with PMF (20 with prefibrotic-PMF) and 13 healthy controls (CTRLs). Levels of constitutive and TPO induced p-STAT5, and IL6 induced p-STAT3 were higher in patients than in CTRLs. Constitutive p-STAT5 values were lower in CALR than homozygous JAK2V617F mutated CD34+ cells from PMF patients. Moreover, constitutive p-STAT5 and IL6 induced p-STAT3 values correlated directly with circulating CD34+ cell number/L, and inversely with the frequency of circulating CD34+ cells expressing CXCR4. Constitutive p-STAT5 values of CD34+ cells were also inversely correlated with hemoglobin levels. When the patients were divided according with presence/absence of JAK2V617F mutation, all the correlations described characterized the JAK2V617F+ patients with prefibrotic-PMF (P-PMF). In conclusion, increased constitutive p-STAT5 and IL6 induced p-STAT3 values in circulating CD34+ cells characterize patients with PMF. Constitutive p-STAT5 and IL6 induced p-STAT3 values correlate with circulating CD34+ cell number/L, the frequency of circulating CD34+ cells expressing CXCR4 and hemoglobin levels within the prefibrotic JAK2V617F+ patient population. Our data point toward a complex activation of STAT5-dependent pathways in the stem/progenitor cell compartment, that characterize the phenotypic diversity of PMF.
- Published
- 2019
- Full Text
- View/download PDF
14. Endothelial TRPV1 as an Emerging Molecular Target to Promote Therapeutic Angiogenesis
- Author
-
Sharon Negri, Pawan Faris, Vittorio Rosti, Maria Rosa Antognazza, Francesco Lodola, and Francesco Moccia
- Subjects
therapeutic angiogenesis ,Ca2+ signaling ,TRPV1 ,vascular endothelial cells ,endothelial colony forming cells ,erythropoietin ,Cytology ,QH573-671 - Abstract
Therapeutic angiogenesis represents an emerging strategy to treat ischemic diseases by stimulating blood vessel growth to rescue local blood perfusion. Therefore, injured microvasculature may be repaired by stimulating resident endothelial cells or circulating endothelial colony forming cells (ECFCs) or by autologous cell-based therapy. Endothelial Ca2+ signals represent a crucial player in angiogenesis and vasculogenesis; indeed, several angiogenic stimuli induce neovessel formation through an increase in intracellular Ca2+ concentration. Several members of the Transient Receptor Potential (TRP) channel superfamily are expressed and mediate Ca2+-dependent functions in vascular endothelial cells and in ECFCs, the only known truly endothelial precursor. TRP Vanilloid 1 (TRPV1), a polymodal cation channel, is emerging as an important player in endothelial cell migration, proliferation, and tubulogenesis, through the integration of several chemical stimuli. Herein, we first summarize TRPV1 structure and gating mechanisms. Next, we illustrate the physiological roles of TRPV1 in vascular endothelium, focusing our attention on how endothelial TRPV1 promotes angiogenesis. In particular, we describe a recent strategy to stimulate TRPV1-mediated pro-angiogenic activity in ECFCs, in the presence of a photosensitive conjugated polymer. Taken together, these observations suggest that TRPV1 represents a useful target in the treatment of ischemic diseases.
- Published
- 2020
- Full Text
- View/download PDF
15. VEGFA rs3025020 Polymorphism Contributes to CALR-Mutation Susceptibility and Is Associated with Low Risk of Deep Vein Thrombosis in Primary Myelofibrosis
- Author
-
Rita Campanelli, Margherita Massa, Adriana Carolei, Laura Villani, Carlotta Abbà, Robert Peter Gale, Paolo Catarsi, G. Barosi, Annalisa de Silvstri, and Vittorio Rosti
- Subjects
medicine.medical_specialty ,business.industry ,Deep vein ,Single-nucleotide polymorphism ,vegfa polymorphism ,medicine.disease ,Thrombosis ,Gastroenterology ,deep vein thrombosis ,Minor allele frequency ,medicine.anatomical_structure ,calr mutation ,Polymorphism (computer science) ,RC666-701 ,Internal medicine ,Genotype ,primary myelofibrosis ,rs3025020 ,medicine ,Diseases of the circulatory (Cardiovascular) system ,Original Article ,Cumulative incidence ,business ,Myelofibrosis - Abstract
Background Single nucleotide polymorphisms (SNPs) in vascular endothelial growth factor A (VEGFA) are associated with susceptibility to several diseases including cancer. Correlations between VEGFA rs3025020 genotypes with clinical and laboratory features of primary myelofibrosis (PMF) are unstudied. Methods DNA was analyzed by real-time polymerase chain reaction for VEGFA rs3025020 genotypes in a cohort of 844 subjects with PMF and in two cohorts of normal subjects (N = 247 and N = 107). Results Frequency of rs3025020 minor allele (T) was not significantly different in subjects with PMF compared with normals; however, the T-allele was more frequent in PMF subjects with a calreticulin (CALR)-mutated genotype compared with normals (35 vs. 27%; OR = 1.47 [95% CI, 1.09, 1.98] p = 0.011), especially in subjects with a CALR-type 2/type 2-like mutation (43 vs. 27%; OR = 2.01 [1.25, 3.24] p = 0.004). CALR mutants with the rs3025020 TT genotype had higher CXCR4 expression on CD34-positive blood cells, and those who carried CT/TT genotypes had lower platelet concentrations compared with other genotypes at diagnosis. Overall, subjects with the rs3025020 CT/TT genotype had a lower cumulative incidence of deep vein thrombosis in typical sites (1.6 vs. 4.2%; OR = 0.37 [0.15, 0.90] p = 0.029) and longer interval from diagnosis to first thrombosis (HR = 0.37 [0.14, 0.95] p = 0.039). Conclusion Persons with PMF and the VEGFA rs3025020 minor T-allele are more likely to have a CALR mutation compared with other somatic driver mutations and lower cumulative incidence and hazard for deep vein thrombosis in typical sites.
- Published
- 2021
- Full Text
- View/download PDF
16. The human amniotic fluid stem cell secretome triggers intracellular Ca 2+ oscillations, NF‐κB nuclear translocation and tube formation in human endothelial colony‐forming cells
- Author
-
Sharon Negri, Carolina Balbi, Pawan Faris, Sveva Bollini, Valentina Balducci, Vittorio Rosti, Ambra Costa, and Francesco Moccia
- Subjects
human amniotic fluid stem cell secretome ,Ca ,Angiogenesis ,NAADP ,2+ ,paracrine therapy ,NF-κB ,InsP3Rs ,endothelial colony-forming cells ,angiogenesis ,chemistry.chemical_compound ,Paracrine signalling ,Ca2+ signalling ,Extracellular ,signalling ,TRPV4 ,tubulogenesis ,Humans ,endothelial colony‐forming cells ,Cells, Cultured ,Secretome ,Tube formation ,Chemistry ,Stem Cells ,NF‐κB ,NF-kappa B ,Endothelial Cells ,Original Articles ,Cell Biology ,Amniotic Fluid ,In vitro ,Cell biology ,Protein Transport ,Culture Media, Conditioned ,Molecular Medicine ,Original Article ,Calcium ,Stem cell ,Intracellular ,Signal Transduction - Abstract
Second trimester foetal human amniotic fluid‐derived stem cells (hAFS) have been shown to possess remarkable cardioprotective paracrine potential in different preclinical models of myocardial injury and drug‐induced cardiotoxicity. The hAFS secretome, namely the total soluble factors released by cells in their conditioned medium (hAFS‐CM), can also strongly sustain in vivo angiogenesis in a murine model of acute myocardial infarction (MI) and stimulates human endothelial colony‐forming cells (ECFCs), the only truly recognized endothelial progenitor, to form capillary‐like structures in vitro. Preliminary work demonstrated that the hypoxic hAFS secretome (hAFS‐CMHypo) triggers intracellular Ca2+ oscillations in human ECFCs, but the underlying mechanisms and the downstream Ca2+‐dependent effectors remain elusive. Herein, we found that the secretome obtained by hAFS undergoing hypoxic preconditioning induced intracellular Ca2+ oscillations by promoting extracellular Ca2+ entry through Transient Receptor Potential Vanilloid 4 (TRPV4). TRPV4‐mediated Ca2+ entry, in turn, promoted the concerted interplay between inositol‐1,4,5‐trisphosphate‐ and nicotinic acid adenine dinucleotide phosphate‐induced endogenous Ca2+ release and store‐operated Ca2+ entry (SOCE). hAFS‐CMHypo‐induced intracellular Ca2+ oscillations resulted in the nuclear translocation of the Ca2+‐sensitive transcription factor p65 NF‐κB. Finally, inhibition of either intracellular Ca2+ oscillations or NF‐κB activity prevented hAFS‐CMHypo‐induced ECFC tube formation. These data shed novel light on the molecular mechanisms whereby hAFS‐CMHypo induces angiogenesis, thus providing useful insights for future therapeutic strategies against ischaemic‐related myocardial injury.
- Published
- 2021
- Full Text
- View/download PDF
17. Clonal Megakaryocyte Dysplasia with Normal Blood Values Is a Distinct Myeloproliferative Neoplasm
- Author
-
Margherita Massa, Vittorio Rosti, Umberto Magrini, Massimo Primignani, Francesco Bertolini, Rita Campanelli, Corrado Lodigiani, Giuliana Gregato, Carlotta Abbà, Laura Villani, Robert Peter Gale, Giovanni Barosi, Adriana Carolei, and Paolo Catarsi
- Subjects
Adult ,Male ,medicine.medical_specialty ,Mutation, Missense ,Megakaryocyte ,Bone Marrow ,Internal medicine ,medicine ,Humans ,Myelofibrosis ,Myeloproliferative neoplasm ,Retrospective Studies ,Myeloproliferative Disorders ,business.industry ,Incidence (epidemiology) ,Thrombosis ,Retrospective cohort study ,Hematology ,General Medicine ,Janus Kinase 2 ,Middle Aged ,medicine.disease ,medicine.anatomical_structure ,Amino Acid Substitution ,Dysplasia ,Hematologic Neoplasms ,Female ,Bone marrow ,Calreticulin ,business ,Megakaryocytes - Abstract
Introduction: In 1991, we reported 18 persons with a clinical-pathologic entity and termed atypical myeloproliferative disorder because they did not meet the contemporary diagnostic criteria for a myeloproliferative neoplasm. We sought to gain further knowledge on this disease entity. Methods: This retrospective cohort study included consecutive subjects registered in the database of the Center for the Study of Myelofibrosis in Pavia, Italy, from 1998 to 2020 (June), and diagnosed with atypical myeloproliferative disorder according to our adjudicated criteria. We studied clinical, histological, cytogenetic, and molecular covariates and risks of thrombosis, disease progression, and death. Data were compared with those of concurrent subjects with prefibrotic myelofibrosis. Results: Fifteen new subjects with atypical myeloproliferative disorder were identified. Seven were male. Median age was 50 years (IQR, 41–54 years). Thirteen were diagnosed with a synchronous symptomatic or incidentally detected thrombotic event. The bone marrow showed megakaryocyte hyperplasia with dysplasia. JAK2V617F was present in 10 subjects and CALR mutation in one. No other somatic mutations were identified in next generation sequencing. After a median follow-up of 101 months (IQR, 40–160 months), no subject had disease progression or blast transformation. Incidence of post-diagnosis or recurrent thrombosis was 3.9 events (95% confidence interval, 3.5–4.0) and 5.0 events (4.6–5.6) per 100 person-years. Features of subjects with atypical myeloproliferative disorder differed markedly from those of 546 subjects with prefibrotic myelofibrosis. Conclusion: Our data indicate that these 15 persons have a distinct myeloproliferative neoplasm. We propose naming this new disorder clonal megakaryocyte dysplasia with normal blood values.
- Published
- 2021
- Full Text
- View/download PDF
18. Preclinical Validation of an Advanced Therapy Medicinal Product Based on Cytotoxic T Lymphocytes Specific for Mutated Nucleophosmin (NPM1mut) for the Treatment of NPM1mut-Acute Myeloid Leukemia
- Author
-
Marica De Cicco, Ivana Lagreca, Sabrina Basso, Patrizia Barozzi, Stella Muscianisi, Alba Bianco, Giovanni Riva, Sara Di Vincenzo, Chiara Pulvirenti, Davide Sapuppo, Mariangela Siciliano, Vittorio Rosti, Anna Candoni, Marco Zecca, Fabio Forghieri, Mario Luppi, and Patrizia Comoli
- Subjects
Cancer Research ,acute myeloid leukemia ,NPM1 mutation ,T cell therapy ,hematopoietic stem cell transplantation ,minimal residual disease ,Oncology - Abstract
Acute myeloid leukemia (AML) with nucleophosmin (NPM1) genetic mutations is the most common subtype in adult patients. Refractory or relapsed disease in unfit patients or after allogeneic hematopoietic stem cell transplantation (allo-HSCT) has a poor prognosis. NPM1-mutated protein, stably expressed on tumor cells but not on normal tissues, may serve as an ideal target for NPM1-mutated AML immunotherapy. The study aim was to investigate the feasibility of producing mutated-NPM1-specific cytotoxic T cells (CTLs) suitable for somatic cell therapy to prevent or treat hematologic relapse in patients with NPM1-mutated AML. T cells were expanded or primed from patient or donor peripheral blood mononuclear cells by NPM1-mutated protein-derived peptides, and tested for leukemia antigen-targeted cytotoxic activity, cytokine production and hematopoietic precursor inhibitory effect. We found that mutated-NPM1-specific CTLs, displaying specific cytokine production and high-level cytotoxicity against patients’ leukemia blasts, and limited inhibitory activity in clonogenic assays, could be obtained from both patients and donors. The polyfunctional mutated-NPM1-specific CTLs included both CD8+ and CD4+ T cells endowed with strong lytic capacity. Our results suggest that mutated-NPM1-targeted CTLs may be a useful therapeutic option to control low-tumor burden relapse following conventional chemotherapy in older NPM1-mutated AML patients or eradicate persistent MRD after HSCT.
- Published
- 2023
- Full Text
- View/download PDF
19. Long-term safety and efficacy of patisiran for hereditary transthyretin-mediated amyloidosis with polyneuropathy: 12-month results of an open-label extension study
- Author
-
David Adams, Michael Polydefkis, Alejandra González-Duarte, Jonas Wixner, Arnt V Kristen, Hartmut H Schmidt, John L Berk, Inés Asunción Losada López, Angela Dispenzieri, Dianna Quan, Isabel M Conceição, Michel S Slama, Julian D Gillmore, Theodoros Kyriakides, Senda Ajroud-Driss, Márcia Waddington-Cruz, Michelle M Mezei, Violaine Planté-Bordeneuve, Shahram Attarian, Elizabeth Mauricio, Thomas H Brannagan, Mitsuharu Ueda, Emre Aldinc, Jing Jing Wang, Matthew T White, John Vest, Erhan Berber, Marianne T Sweetser, Teresa Coelho, Giuseppe Vita, Vincenzo Rizzo, Massimo Russo, Anna Mazzeo, Luca Gentile, Caitlin Brueckner, Victoria Lazzari, Janice Wiesman, Douglas DeLong, Jennifer Victory, James Dalton, John May, Catherine Gilmore, Saran Diallo, Emilien Delmont, Jean Pouget, Annie Verschueren, Aude-Marie Grapperon, Emmanuelle Campana-Salort, Ana Lopes, Filipa Lamas, Carlos Neves, Jose Castro, Pedro Pereira, Isabel Castro, Ana Franco, Miguel Oliveira Santos, Conceição de Azevedo Coutinho, Catarina Falcao de Campos, Antonio Hipólito Reis, Nuno Correia, Javier M Perez, Ana Martins da Silva, Cristina Alves, Marcio Cardoso, Katia Valdrez, Julia R Monte, Bernardete Pessoa, Nadia Guimaraes, Monica Freitas, Joana Ramalho, Natalia Ferreira, Daisuke Kuzume, Celine Tard, Nawal Waucquier, Isabelle Rougeaux, Sylvie Brice, Emmanuelle Kasprzyk, Elise Elrezzi, Sayah Meguig, Eric Hachulla, Clement Gauvain, Maria-Claire Migaud-Chervy, Dominique Deplanque, Elsa Jozefowicz, Loic Lebellec, Line Balaya-Gouraya, Nathalie Jehan Lacour, Halima Bournane, Nathalie Martin, Mongia Elabed, Niamey Sacko, Yasmine Boubrit, Amina Gaouar, Fetra Rakotondratafika, Marie Théaudin-Saliou, Cécile Cauquil-Michon, Celine Labeyrie, Adeline Not, Abdallah Al-Salameh, Anne-Lise Lecoq, Maeva Stephant, Andoni Echaniz-Laguna, Laurent Becquemont, Guillemette Beaudonnet, Vincent Algalarrondo, Ludivine Eliahou, Antoine Rousseau, Aissatou Signate, Emeline Berthelot, Jocelyn Inamo, Laetitia Vervoitte, Cecile Focseneanu, Thierry Gendre, Raphaele Arrouasse, Samar S. Ayache, Laura Ernande, Philippe Le Corvoisier, Hayet Salhi, Ariane Choumert, Vincent Ehinger, Julie Ruiz, Cyril Charlin, Thomas Megelin, Thomas H Brannagan III, Raisy Fayerman, Arreum Kim, Allan Paras, Leidy J Gonzalez, Steven Tsang, Fernanda Wajnsztajn, Jeffrey Shije, Christina Ulane, Inna Kleyman, Louis Weimer, Comana Cioroiu, Sakis Lambrianides, Rana Abu-Manneh, Eleni Zamba-Papanicolaou, Petros Agathangelou, Eleni Leonidou, Satoshi Tada, Akemi Fujita, Masahiro Nagai, Rina Ando, Yuko Hosokawa, Yuki Yamanishi, J. Scott Overcash, Elena Giardino, Leslie Boyer, Lien Dang, An Le, Tyler Nguyen, Lien Giang, Peter Sellers, Leyla Tran, Nghi Truong, Maita Vinas, Nicole Hrkman, Sarah Miller, David Nguyen, Ashley Smith, Helen Pu, Steve Li, Thao Vuong, Holly Dioso, Sinikka Green, Kia Lee, Hanh Chu, Michael Waters, Derya J Coskun, Karla A Zepeda, William O'Riordan, Laura Obici, Andrea Cortese, Alessandro Lozza, Giampaolo Merlini, Vittorio Rosti, Mario Sabatelli, Giulia Bisogni, Daniela Bernardo, Marco Luigetti, Andrea Di Paolantonio, Valeria Guglielmino, Angela Romano, Hans Nienhuis, Janita Bulthuis-Kuiper, Olga Gerk, Hannah Ulbricht, Lenka Taylor, Eva Meyle, Natalia Kleinschmidt, David Meyrath, Simone Noe-Schwenn, Ulrike Meng, Ralf Bauer, Fabian aus dem Siepen, Selina Hein, Tetsuya Takahashi, Tomohiko Oshita, Yoko Koujin, Shuichiro Neshige, Tomohisa Nezu, Akiko Segawa, Hiroki Ueno, Hiroyuki Morino, Josep M Campistol, Lida Maria Rodas Marin, Josep Miquel Blasco Pelicano, Lucía Galán Dávila, Marta Palacios, Vanesa Pytel Cordoba, Antonio Guerrero Sola, Alejandro Horga, Julián García Feijoo, Leopoldo Perez de Isla, Wilson Marques Júnior, Mariana Moscardini, Debora Cristina Litcanov, Ana Flavia Viera Lima, Leonardo Rodrigues, Barbara Marques Coutinho, Carolina Lavigne Moreira, Vanessa Daccach Marques, Francisco Munoz Beamud, Álvaro Gragera Martínez, Cristina Borrachero, Eugenia Cisneros Barroso, Adrián Rodríguez Rodríguez, Monica Sanz, Elena Rigo Oliver, Juan González Moreno, Jose M Gamez Martinez, Cristina Descals, Mercedes Uson, Francisco Jose Vega, Antoni Figuerola, Carles Montala, Moises Dias da Silva, Renata Gervais de Santa Rosa, Luiz Felipe Pinto, Marcus Vinicius Pinto, Amanda Cardoso Berensztejn, Fabio Barroso, Andrea Lautre, Lucas G Orellana, Maria Alejandra González-Duarte Briseño, Karla Cárdenas-Soto, Brenda Poled Jiménez López, Sandra Lorena Pérez-Castañeda, Carlos Gerardo Cantú Brito, David Rivera de la Parra, Jose Pablo Hernandez Reyes, Maria del Mar Saniger Alba, Elia Criollo Mora, Yesim Parman, Kus Jülide Rezzan, Erdi Sahin, Nail G Serbest, Hacer Durmus, Arman Cakar, Nuriye Ilknur Tugal Tutkun, Sacit Karamursel, Ali Elitok, Nermin G Sirin Inan, Emre Altinkurt, Jing Ye, Adriane C Allen, Vinay Chaudhry, Raquel Jarrett, Neil Bressler, Kathleen L Burks, Qingfeng Liu, Mohammad Khoshnoodi, Daniel P Judge, Geno Vista, Syed Mahmood Shah, Hirotoshi Hamaguchi, Junko Oda, Emi Fukase, Ikuko Taniguchi, Tetsuya Oda, Hironobu Endo, Masahiro Shimomura, Kimitaka Katanazaka, Shusuke Koto, Takahiro Nakano, Christof Scheid, Andreas Zueiter, Lars Pester, Doreen Walter, Betül Özdemir, Lukas F Frenzel, Udo Holtick, Jeeyoung Oh, Hee Jin Kim, Hyun Jin Shin, Kyomin Choi, Taro Yamashita, Teruaki Masuda, Yohei Misumi, Akihiko Ueda, Keiichi Nakahara, Akiko Yorita, Seiko Tsuruhisa, Takayuki Taniwaki, Masaya Harada, Taiga Moritaka, Naonori Sakurada, Elizabeth A Mauricio, Amber Baskin, Elliot Dimberg, Amie Fonder, Miriam Hobbs, Stephen J Russell, Peter Dyck, Wilson Gonsalves, Nelson Leung, Thomas E Witzig, Steven R Zeldenrust, Lisa Hwa, Prashant Kapoor, Shaji K Kumar, Yi Lin, John A Lust, Vincent S Rajkumar, David Dingli, Morie A Gertz, Ronald Go, Suzanne R Hayman, Samir Dalia, Esmeralda Carrillo, Peter Gorevic, Garnette Mason, Chi-Chao Chao, Ming-Jen Lee, Jen-Jen Su, Sung-Tsang Hsieh, Li-Kai Tsai, Shin-Joe Yeh, Chih-Chao Yang, Senda Ajroud-Driss Ajroud-Driss, Patricia Casey, Benjamin C Joslin, Miriam Freimer, Alison Sankey, Amanda Kenepp, Sarah Heintzman, Samantha LoRusso, Youichi Hokezu, Byoung-Joon Kim, JuHyeon Kim, Ga Yeon Lee, Eun Bin Cho, Eun-Seok Jeon, Ju-Hong Min, Jin Myoung Seok, Hye Lim Lee, Jae Hong Park, Yoshiki Sekijima, Chinatsu Miyazawa, Nagaaki Kato, Dai Kishida, Akiyo Hineno, Minori Kodaira, Tsuneaki Yoshinaga, Teruyoshi Miyahara, Akira Imai, Kazuhiko Matsumoto, Kon-Ping Lin, Yi-Chung Lee, Malin Falk, Bjorn Pilebro, Ole Suhr, Per Lindqvist, Karin Soderberg, Fatima Pedrosa-Domellöf, Intissar Anan, Erik Nordh, Ivaylo Tournev, Sashka Zhelyazkova-Glaveeva, Zheyna Cherneva, Staiko Sarafov, Teodora Chamova, Sylvia Cherninkova-Gopina, Frauke Friebel, Andree Zibert, Natasa Mihailovic, Friederike Schubert, Elena Vorona, Larissa Lahme, Anna Huesing-Kabar, Matthias Schilling, Iyad Kabar, Ana Martinez-Naharro, Liza Chacko, Oliver Cohen, Steven Law, Tamer Rezk, Helen J Lachmann, Brianna Blume, Stacy Dixon, Soon Chai Low, Soo Looi Chan, He Eng Li Lim, Khean Jin Goh, Deborah Kraus, Kristin Jack, N. Kevin Wade, Glenn Lopate, Brittany Zwijack, Julaine Florence, R. Brian Sommerville, Graeme Stewart, Julie Ryder, Linda Mekhael, Mark Taylor, Daniel Suan, Karen Wells, Paula Stone, Amenze Itoya, Mercy Owusu-Sekyere, Desmond Thai, Ilonah Chahine, Salve Pedrosa, Thi Hoa (Therese) Do, and Repositório da Universidade de Lisboa
- Subjects
Adult ,Male ,medicine.medical_specialty ,Drug-Related Side Effects and Adverse Reactions ,030204 cardiovascular system & hematology ,Placebo ,Severity of Illness Index ,Polyneuropathies ,03 medical and health sciences ,0302 clinical medicine ,Quality of life ,Internal medicine ,Outcome Assessment, Health Care ,Severity of illness ,medicine ,Humans ,Prealbumin ,RNA, Small Interfering ,Infusions, Intravenous ,Adverse effect ,Aged ,Amyloid Neuropathies, Familial ,business.industry ,Middle Aged ,medicine.disease ,Interim analysis ,amyloidosis, transthyretin, polyneuropathy, patisiran, OLE study ,Clinical trial ,Female ,Neurology (clinical) ,business ,Polyneuropathy ,030217 neurology & neurosurgery ,Progressive disease - Abstract
© 2020 Elsevier Ltd. All rights reserved., Background: Hereditary transthyretin-mediated amyloidosis is a rare, inherited, progressive disease caused by mutations in the transthyretin (TTR) gene. We assessed the safety and efficacy of long-term treatment with patisiran, an RNA interference therapeutic that inhibits TTR production, in patients with hereditary transthyretin-mediated amyloidosis with polyneuropathy. Methods: This multicentre, open-label extension (OLE) trial enrolled patients at 43 hospitals or clinical centres in 19 countries as of Sept 24, 2018. Patients were eligible if they had completed the phase 3 APOLLO or phase 2 OLE parent studies and tolerated the study drug. Eligible patients from APOLLO (patisiran and placebo groups) and the phase 2 OLE (patisiran group) studies enrolled in this global OLE trial and received patisiran 0·3 mg/kg by intravenous infusion every 3 weeks with plans to continue to do so for up to 5 years. Efficacy assessments included measures of polyneuropathy (modified Neuropathy Impairment Score +7 [mNIS+7]), quality of life, autonomic symptoms, nutritional status, disability, ambulation status, motor function, and cardiac stress, with analysis by study groups (APOLLO-placebo, APOLLO-patisiran, phase 2 OLE patisiran) based on allocation in the parent trial. The global OLE is ongoing with no new enrolment, and current findings are based on the interim analysis of the patients who had completed 12-month efficacy assessments as of the data cutoff. Safety analyses included all patients who received one or more dose of patisiran up to the data cutoff. This study is registered with ClinicalTrials.gov, NCT02510261. Findings: Between July 13, 2015, and Aug 21, 2017, of 212 eligible patients, 211 were enrolled: 137 patients from the APOLLO-patisiran group, 49 from the APOLLO-placebo group, and 25 from the phase 2 OLE patisiran group. At the data cutoff on Sept 24, 2018, 126 (92%) of 137 patients from the APOLLO-patisiran group, 38 (78%) of 49 from the APOLLO-placebo group, and 25 (100%) of 25 from the phase 2 OLE patisiran group had completed 12-month assessments. At 12 months, improvements in mNIS+7 with patisiran were sustained from parent study baseline with treatment in the global OLE (APOLLO-patisiran mean change -4·0, 95 % CI -7·7 to -0·3; phase 2 OLE patisiran -4·7, -11·9 to 2·4). Mean mNIS+7 score improved from global OLE enrolment in the APOLLO-placebo group (mean change from global OLE enrolment -1·4, 95% CI -6·2 to 3·5). Overall, 204 (97%) of 211 patients reported adverse events, 82 (39%) reported serious adverse events, and there were 23 (11%) deaths. Serious adverse events were more frequent in the APOLLO-placebo group (28 [57%] of 49) than in the APOLLO-patisiran (48 [35%] of 137) or phase 2 OLE patisiran (six [24%] of 25) groups. The most common treatment-related adverse event was mild or moderate infusion-related reactions. The frequency of deaths in the global OLE was higher in the APOLLO-placebo group (13 [27%] of 49), who had a higher disease burden than the APOLLO-patisiran (ten [7%] of 137) and phase 2 OLE patisiran (0 of 25) groups. Interpretation: In this interim 12-month analysis of the ongoing global OLE study, patisiran appeared to maintain efficacy with an acceptable safety profile in patients with hereditary transthyretin-mediated amyloidosis with polyneuropathy. Continued long-term follow-up will be important for the overall assessment of safety and efficacy with patisiran.
- Published
- 2021
- Full Text
- View/download PDF
20. Long Term Follow-up of an Investigator-Initiated Phase 2 Study of Ruxolitinib in MPN-Associated Splancnic Vein Thrombosis (SVT-RUXO)
- Author
-
Chiara Paoli, Paola Guglielmelli, Francesco Mannelli, Miriam Borella, Vittorio Rosti, Elisa Rumi, Maria Chiara Finazzi, Oscar Borsani, Carmela Mannarelli, Maria Esposito, Mario Xhani, Lisa Pieri, Tiziano Barbui, Alessandro Rambaldi, and Alessandro Vannucchi
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
21. Cells coexpressing both myeloid and endothelial markers are detectable in the spleen and bone marrow of patients with primary myelofibrosis
- Author
-
Rita Campanelli, Carlotta Abbà, Adriana Carolei, Paolo Catarsi, Giovanni Barosi, Margherita Massa, and Vittorio Rosti
- Subjects
Cancer Research ,Hemangioblasts ,Antigens, CD34 ,Bone Marrow Cells ,Cell Differentiation ,Cell Biology ,Hematology ,Bone Marrow ,Primary Myelofibrosis ,Genetics ,Humans ,Molecular Biology ,Spleen ,Biomarkers - Abstract
Different bodies of evidence support the existence of a common origin of hematopoietic and endothelial lineages; moreover, recent studies have indicated the presence of a hemogenic endothelium and a common hemato-endothelial precursor both in the embryo and in the cord blood. Conversely, to our knowledge, there is no evidence of such bipotential cells in human postnatal tissues or blood. In this study, we investigated the presence and phenotype of "transitional" cells in different tissues of patients with primary myelofibrosis (PMF). Using confocal microscopy and flow cytometry, we identified a rare cell population in the bone marrow and spleen of patients with PMF, which coexpresses the endothelial marker CD144 (vascular endothelial (VE)-cadherin), the pan-hematopoietic marker CD45, the early myeloid marker CD33, and CD34, a common endothelial and hematopoietic antigen.
- Published
- 2022
22. Ruxolitinib versus best available therapy in patients with polycythemia vera: 80-week follow-up from the RESPONSE trial
- Author
-
Srdan Verstovsek, Alessandro M. Vannucchi, Martin Griesshammer, Tamas Masszi, Simon Durrant, Francesco Passamonti, Claire N. Harrison, Fabrizio Pane, Pierre Zachee, Keita Kirito, Carlos Besses, Masayuki Hino, Beatriz Moiraghi, Carole B. Miller, Mario Cazzola, Vittorio Rosti, Igor Blau, Ruben Mesa, Mark M. Jones, Huiling Zhen, Jingjin Li, Nathalie Francillard, Dany Habr, and Jean-Jacques Kiladjian
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
RESPONSE is an open-label phase 3 study evaluating the Janus kinase 1/Janus kinase 2 inhibitor ruxolitinib versus best available therapy for efficacy/safety in hydroxyurea-resistant or intolerant patients with polycythemia vera. This preplanned analysis occurred when all patients completed the Week 80 visit or discontinued. Objectives included evaluating the durability of the primary response (Week 32 phlebotomy-independent hematocrit control plus ≥35% spleen volume reduction), its components, and that of complete hematologic remission; and long-term safety. Median exposure was 111 weeks; 91/110 (82.7%) patients randomized to ruxolitinib remained on treatment. No patients continued best available therapy (98/112 [87.5%] crossed over to ruxolitinib, most at/soon after Week 32). At Week 32, primary response was achieved by 22.7% vs. 0.9% of patients randomized to ruxolitinib and best available therapy, respectively (hematocrit control, 60.0% vs. 18.8%; spleen response, 40.0% vs. 0.9%). The probability of maintaining primary and hematocrit responses for ≥80 weeks was 92% and 89%, respectively; 43/44 spleen responses were maintained until Week 80. Complete hematologic remission at Week 32 was achieved in 23.6% of ruxolitinib-randomized patients; the probability of maintaining complete hematologic remission for ≥80 weeks was 69%. Among ruxolitinib crossover patients, 79.2% were not phlebotomized, and 18.8% achieved a ≥35% reduction from baseline in spleen volume after 32 weeks of treatment. New or worsening hematologic laboratory abnormalities in ruxolitinib-treated patients were primarily grade 1/2 decreases in hemoglobin, lymphocytes, and platelets. The thromboembolic event rate per 100 patient-years was 1.8 with randomized ruxolitinib treatment vs. 8.2 with best available therapy. These data support ruxolitinib as an effective long-term treatment option for hydroxyurea-resistant or intolerant patients with polycythemia vera. This trial was registered at clinicaltrials.gov identifier: 01243944.
- Published
- 2016
- Full Text
- View/download PDF
23. Tie2 Expressing Monocytes in the Spleen of Patients with Primary Myelofibrosis.
- Author
-
Rita Campanelli, Gabriela Fois, Paolo Catarsi, Valentina Poletto, Laura Villani, Benedetta Gaia Erba, Luigi Maddaluno, Basilio Jemos, Silvia Salmoiraghi, Paola Guglielmelli, Vittorio Abbonante, Christian Andrea Di Buduo, Alessandra Balduini, Alessandra Iurlo, Giovanni Barosi, Vittorio Rosti, Margherita Massa, and AGIMM Investigators
- Subjects
Medicine ,Science - Abstract
Primary myelofibrosis (PMF) is a Philadelphia-negative (Ph-) myeloproliferative disorder, showing abnormal CD34+ progenitor cell trafficking, splenomegaly, marrow fibrosis leading to extensive extramedullary haematopoiesis, and abnormal neoangiogenesis in either the bone marrow or the spleen. Monocytes expressing the angiopoietin-2 receptor (Tie2) have been shown to support abnormal angiogenic processes in solid tumors through a paracrine action that takes place in proximity to the vessels. In this study we investigated the frequency of Tie2 expressing monocytes in the spleen tissue samples of patients with PMF, and healthy subjects (CTRLs), and evaluated their possible role in favouring spleen angiogenesis. We show by confocal microscopy that in the spleen tissue of patients with PMF, but not of CTRLs, the most of the CD14+ cells are Tie2+ and are close to vessels; by flow cytometry, we found that Tie2 expressing monocytes were Tie2+CD14lowCD16brightCDL62-CCR2- (TEMs) and their frequency was higher (p = 0.008) in spleen tissue-derived mononuclear cells (MNCs) of patients with PMF than in spleen tissue-derived MNCs from CTRLs undergoing splenectomy for abdominal trauma. By in vitro angiogenesis assay we evidenced that conditioned medium of immunomagnetically selected spleen tissue derived CD14+ cells of patients with PMF induced a denser tube like net than that of CTRLs; in addition, CD14+Tie2+ cells sorted from spleen tissue derived single cell suspension of patients with PMF show a higher expression of genes involved in angiogenesis than that found in CTRLs. Our results document the enrichment of Tie2+ monocytes expressing angiogenic genes in the spleen of patients with PMF, suggesting a role for these cells in starting/maintaining the pathological angiogenesis in this organ.
- Published
- 2016
- Full Text
- View/download PDF
24. Co-mutation pattern, clonal hierarchy, and clone size concur to determine disease phenotype of SRSF2P95-mutated neoplasms
- Author
-
Maria Creignou, Anna Gallì, Paola Guglielmelli, Mario Cazzola, Johanna Ungerstedt, Seishi Ogawa, Silvia Catricalà, Ettore Rizzo, Marios Dimitriou, Eva Hellström-Lindberg, Elisa Bono, Alessandro M. Vannucchi, Martina Sarchi, Elisa Rumi, Luca Malcovati, Vittorio Rosti, Yasuhito Nannya, Marco Roncador, Daniela Pietra, Chiara Elena, Elisabetta Molteni, and Gabriele Todisco
- Subjects
0301 basic medicine ,Genetics ,Cancer Research ,education.field_of_study ,Myeloid ,Genetic heterogeneity ,Myelodysplastic syndromes ,Population ,Myeloid leukemia ,Hematology ,Biology ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Monocytosis ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,medicine ,education ,Myelofibrosis ,Dominance (genetics) - Abstract
Somatic mutations in splicing factor genes frequently occur in myeloid neoplasms. While SF3B1 mutations are associated with myelodysplastic syndromes (MDS) with ring sideroblasts, SRSF2P95 mutations are found in different disease categories, including MDS, myeloproliferative neoplasms (MPN), myelodysplastic/myeloproliferative neoplasms (MDS/MPN), and acute myeloid leukemia (AML). To identify molecular determinants of this phenotypic heterogeneity, we explored molecular and clinical features of a prospective cohort of 279 SRSF2P95-mutated cases selected from a population of 2663 patients with myeloid neoplasms. Median number of somatic mutations per subject was 3. Multivariate regression analysis showed associations between co-mutated genes and clinical phenotype, including JAK2 or MPL with myelofibrosis (OR = 26.9); TET2 with monocytosis (OR = 5.2); RAS-pathway genes with leukocytosis (OR = 5.1); and STAG2, RUNX1, or IDH1/2 with blast phenotype (MDS or AML) (OR = 3.4, 1.9, and 2.1, respectively). Within patients with SRSF2-JAK2 co-mutation, JAK2 dominance was invariably associated with clinical feature of MPN, whereas SRSF2 mutation was dominant in MDS/MPN. Within patients with SRSF2-TET2 co-mutation, clinical expressivity of monocytosis was positively associated with co-mutated clone size. This study provides evidence that co-mutation pattern, clone size, and hierarchy concur to determine clinical phenotype, tracing relevant genotype-phenotype associations across disease entities and giving insight on unaccountable clinical heterogeneity within current WHO classification categories.
- Published
- 2020
- Full Text
- View/download PDF
25. Cytogenetic study in primary myelofibrosis at diagnosis: Clinical and histological association and impact on survival according to WHO 2017 classification in an Italian multicenter series
- Author
-
Anna Bossi, Giuseppe Auteri, Mariarita Sciumè, Giorgio Alberto Croci, Silvana Guerneri, Filippo Brioschi, Ivan Cortinovis, Vittorio Rosti, Umberto Gianelli, Donatella Vincelli, Cristina Bucelli, Bruno Martino, Daniele Cattaneo, Elena Maria Elli, Elena Sabattini, Giuseppe Isimbaldi, Francesca Palandri, Luca Baldini, and Alessandra Iurlo
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Anemia ,Abnormal Karyotype ,Bone marrow fibrosis ,Gastroenterology ,Disease-Free Survival ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Bone Marrow ,Risk Factors ,Lactate dehydrogenase ,Internal medicine ,medicine ,Humans ,Myelofibrosis ,Aged ,Series (stratigraphy) ,business.industry ,Karyotype ,Hematology ,General Medicine ,Middle Aged ,Prognosis ,medicine.disease ,Survival Rate ,Italy ,Oncology ,chemistry ,Primary Myelofibrosis ,International Prognostic Scoring System ,030220 oncology & carcinogenesis ,Cytogenetic Analysis ,Cohort ,Female ,business ,Follow-Up Studies ,030215 immunology - Abstract
We analyzed cytogenetic data at diagnosis in 395 primary myelofibrosis (PMF) patients to evaluate any possible association between karyotype and WHO 2017 classification and its impact on prognosis. All the cases were diagnosed and followed at five Italian Hematological Centers between November 1983 and December 2016. An abnormal karyotype (AK) was found in 69 patients and clustered differently according to bone marrow fibrosis grade as it was found in 31 (27.0%) cases with overt fibrotic and 38 (13.6%) with pre-fibrotic PMF (p = 0.001). Sex, anemia, thrombocytopenia, circulating blasts ≥1%, higher lactate dehydrogenase, and International Prognostic Scoring System risk classes were all significantly associated with karyotype. At a median follow-up of >6 years, 101 deaths were recorded. Survival was different between AK and normal karyotype (NK) patients with an estimated median overall survival (OS) of 11.6 and 25.7 years, respectively (p = 0.0148). In conclusion, in our cohort around 20% of patients had an AK, more frequently in subjects with an advanced bone marrow fibrosis grade and clinical-laboratory features indicative of a more aggressive disease. This study shows that an AK confers a more severe clinical phenotype and impacts adversely on OS, thus representing an additional parameter to be considered in the evaluation of PMF prognosis.
- Published
- 2020
- Full Text
- View/download PDF
26. Reduced CXCR4-expression on CD34-positive blood cells predicts outcomes of persons with primary myelofibrosis
- Author
-
Paolo Catarsi, Umberto Magrini, Rita Campanelli, Vittorio Rosti, Laura Villani, Robert Peter Gale, Carlotta Abbà, Margherita Massa, Giovanni Barosi, and Adriana Carolei
- Subjects
Male ,0301 basic medicine ,Receptors, CXCR4 ,Cancer Research ,medicine.medical_specialty ,CD34 ,Antigens, CD34 ,Gastroenterology ,CXCR4 ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Polycythemia vera ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Myelofibrosis ,Aged ,Blood Cells ,Leukopenia ,Essential thrombocythemia ,business.industry ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Survival Rate ,030104 developmental biology ,Oncology ,Primary Myelofibrosis ,International Prognostic Scoring System ,Case-Control Studies ,030220 oncology & carcinogenesis ,Mutation ,Female ,Hemoglobin ,medicine.symptom ,business ,Biomarkers ,Signal Transduction - Abstract
The expression of the CXCR4 chemokine receptor on CD34-positive blood cells is reduced in persons with primary myelofibrosis (PMF). We analyzed the relevance of cytofluorimetric assessment of the percentage of CD34-positive blood cells that had a positive CXCR4 surface expression (CD34/CXCR4-se) in a large cohort of subjects with myeloproliferative neoplasms. Mean CD34/CXCR4-se was lower in subjects with PMF compared with those with essential thrombocythemia (ET) or polycythemia vera (PV). A cutoff value of 39% was associated with a diagnosis of pre-fibrotic PMF vs. ET with a positive predictive value of 97%. In PMF male sex, older age, and MPL mutation were independent correlates of reduced CD34/CXCR4-se and associated with a briefer interval to development of severe anemia, large splenomegaly, thrombocytopenia, leukopenia, elevated CD34-positive blood cells, blast transformation and death. We constructed a prognostic model including age >65 years, hemoglobin 50 × 106/L, and CD34/CXCR4-se
- Published
- 2020
- Full Text
- View/download PDF
27. Long-term efficacy and safety of ruxolitinib versus best available therapy in polycythaemia vera (RESPONSE): 5-year follow up of a phase 3 study
- Author
-
Elisa Rumi, Tamás Masszi, Ruben A. Mesa, Jean-Jacques Kiladjian, Francesco Passamonti, Tuochuan Dong, Nathalie Francillard, Vittorio Rosti, Srdan Verstovsek, Beatriz Moiraghi, Carlos Besses, Simon Durrant, Masayuki Hino, Pierre Zachee, Fabrizio Pane, Claire N. Harrison, Martin Griesshammer, Alessandro M. Vannucchi, Monika Wroclawska, Keita Kirito, Carole B. Miller, Igor Wolfgang Blau, Kiladjian, J. -J., Zachee, P., Hino, M., Pane, F., Masszi, T., Harrison, C. N., Mesa, R., Miller, C. B., Passamonti, F., Durrant, S., Griesshammer, M., Kirito, K., Besses, C., Moiraghi, B., Rumi, E., Rosti, V., Blau, I. W., Francillard, N., Dong, T., Wroclawska, M., Vannucchi, A. M., and Verstovsek, S.
- Subjects
Ruxolitinib ,Time Factors ,Polyethylene Glycol ,Polyethylene Glycols ,0302 clinical medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Hydroxyurea ,Medicine ,Polycythemia Vera ,education.field_of_study ,Fibrinolytic Agent ,Pipobroman ,Hematology ,Recombinant Protein ,Prognosis ,Recombinant Proteins ,Survival Rate ,030220 oncology & carcinogenesis ,Drug Therapy, Combination ,Human ,medicine.drug ,medicine.medical_specialty ,Polycythaemia ,Prognosi ,Population ,Interferon alpha-2 ,Antiviral Agents ,Article ,Follow-Up Studie ,03 medical and health sciences ,Pharmacotherapy ,Fibrinolytic Agents ,Internal medicine ,Nitriles ,Humans ,Adverse effect ,education ,Survival rate ,Antiviral Agent ,Antineoplastic Combined Chemotherapy Protocol ,business.industry ,Quinazoline ,Interferon-alpha ,Anagrelide ,medicine.disease ,Pyrimidines ,Pyrazole ,Quinazolines ,Pyrazoles ,business ,Follow-Up Studies ,030215 immunology - Abstract
Summary Background Polycythaemia vera is a myeloproliferative neoplasm characterised by excessive proliferation of erythroid, myeloid, and megakaryocytic components in the bone marrow due to mutations in the Janus kinase 2 (JAK2) gene. Ruxolitinib, a JAK 1 and JAK 2 inhibitor, showed superiority over best available therapy in a phase 2 study in patients with polycythaemia vera who were resistant to or intolerant of hydroxyurea. We aimed to compare the long-term safety and efficacy of ruxolitinib with best available therapy in patients with polycythaemia vera who were resistant to or intolerant of hydroxyurea. Methods We report the 5-year results for a randomised, open-label, phase 3 study (RESPONSE) that enrolled patients at 109 sites across North America, South America, Europe, and the Asia-Pacific region. Patients (18 years or older) with polycythaemia vera who were resistant to or intolerant of hydroxyurea were randomly assigned 1:1 to receive either ruxolitinib or best available therapy. Patients randomly assigned to the ruxolitinib group received the drug orally at a starting dose of 10 mg twice a day. Single-agent best available therapy comprised hydroxyurea, interferon or pegylated interferon, pipobroman, anagrelide, approved immunomodulators, or observation without pharmacological treatment. The primary endpoint, composite response (patients who achieved both haematocrit control without phlebotomy and 35% or more reduction from baseline in spleen volume) at 32 weeeks was previously reported. Patients receiving best available therapy could cross over to ruxolitinib after week 32. We assessed the durability of primary composite response, complete haematological remission, overall clinicohaematological response, overall survival, patient-reported outcomes, and safety after 5-years of follow-up. This study is registered with ClinicalTrials.gov , NCT01243944 . Findings We enrolled patients between Oct 27, 2010, and Feb 13, 2013, and the study concluded on Feb 9, 2018. Of 342 individuals screened for eligibility, 222 patients were randomly assigned to receive ruxolitinib (n=110, 50%) or best available therapy (n=112, 50%). The median time since polycythaemia vera diagnosis was 8·2 years (IQR 3·9–12·3) in the ruxolitinib group and 9·3 years (4·9–13·8) in the best available therapy group. 98 (88%) of 112 patients initially randomly assigned to best available therapy crossed over to receive ruxolitinib and no patient remained on best available therapy after 80 weeks of study. Among 25 primary responders in the ruxolitinib group, six had progressed at the time of final analysis. At 5 years, the probability of maintaining primary composite response was 74% (95% CI 51–88). The probability of maintaining complete haematological remission was 55% (95% CI 32–73) and the probability of maintaining overall clinicohaematological responses was 67% (54–77). In the intention-to-treat analysis not accounting for crossover, the probability of survival at 5 years was 91·9% (84·4–95·9) with ruxolitinib therapy and 91·0% (82·8–95·4) with best available therapy. Anaemia was the most common adverse event in patients receiving ruxolitinib (rates per 100 patient-years of exposure were 8·9 for ruxolitinib and 8·8 for the crossover population), though most anaemia events were mild to moderate in severity (grade 1 or 2 anaemia rates per 100 patient-years of exposure were 8·0 for ruxolitinib and 8·2 for the crossover population). Non-haematological adverse events were generally lower with long-term ruxolitinib treatment than with best available therapy. Thromboembolic events were lower in the ruxolitinib group than the best available therapy group. There were two on-treatment deaths in the ruxolitinib group. One of these deaths was due to gastric adenocarcinoma, which was assessed by the investigator as related to ruxolitinib treatment. Interpretation We showed that ruxolitinib is a safe and effective long-term treatment option for patients with polycythaemia vera who are resistant to or intolerant of hydroxyurea. Taken together, ruxolitinib treatment offers the first widely approved therapeutic alternative for this post-hydroxyurea patient population. Funding Novartis Pharmaceuticals Corporation.
- Published
- 2020
- Full Text
- View/download PDF
28. Defective interaction of mutant calreticulin and SOCE in megakaryocytes from patients with myeloproliferative neoplasms
- Author
-
Vittorio Abbonante, Vittorio Rosti, Mario Cazzola, Daniela Pietra, Isabelle Plo, Paolo M. Soprano, Christian A. Di Buduo, Alessandra Iurlo, Giovanni Barosi, Daniele Cattaneo, Francesco Moccia, Caroline Marty, Umberto Gianelli, Dmitry Lim, Elisa Rumi, and Alessandra Balduini
- Subjects
Immunology ,Protein Disulfide-Isomerases ,Gene mutation ,medicine.disease_cause ,Biochemistry ,medicine ,Humans ,Stromal Interaction Molecule 1 ,Thrombopoietin ,Myeloproliferative neoplasm ,Mutation ,Myeloproliferative Disorders ,biology ,Essential thrombocythemia ,Chemistry ,STIM1 ,Cell Biology ,Hematology ,medicine.disease ,Calcium Release Activated Calcium Channels ,Store-operated calcium entry ,Neoplasm Proteins ,Case-Control Studies ,biology.protein ,Cancer research ,Calreticulin ,Megakaryocytes ,BLOOD Commentary - Abstract
Approximately one-fourth of patients with essential thrombocythemia or primary myelofibrosis carry a somatic mutation of the calreticulin gene (CALR), the gene encoding for calreticulin. A 52-bp deletion (type I mutation) and a 5-bp insertion (type II mutation) are the most frequent genetic lesions. The mechanism(s) by which a CALR mutation leads to a myeloproliferative phenotype has been clarified only in part. We studied the interaction between calreticulin and store-operated calcium (Ca2+) entry (SOCE) machinery in megakaryocytes (Mks) from healthy individuals and from patients with CALR-mutated myeloproliferative neoplasms (MPNs). In Mks from healthy subjects, binding of recombinant human thrombopoietin to c-Mpl induced the activation of signal transducer and activator of transcription 5, AKT, and extracellular signal-regulated kinase 1/2, determining inositol triphosphate–dependent Ca2+ release from the endoplasmic reticulum (ER). This resulted in the dissociation of the ER protein 57 (ERp57)-mediated complex between calreticulin and stromal interaction molecule 1 (STIM1), a protein of the SOCE machinery that leads to Ca2+ mobilization. In Mks from patients with CALR-mutated MPNs, defective interactions between mutant calreticulin, ERp57, and STIM1 activated SOCE and generated spontaneous cytosolic Ca2+ flows. In turn, this resulted in abnormal Mk proliferation that was reverted using a specific SOCE inhibitor. In summary, the abnormal SOCE regulation of Ca2+ flows in Mks contributes to the pathophysiology of CALR-mutated MPNs. In perspective, SOCE may represent a new therapeutic target to counteract Mk proliferation and its clinical consequences in MPNs.
- Published
- 2020
- Full Text
- View/download PDF
29. Clonal Megakaryocyte Dysplasia with Isolated Thrombocytosis Is a Distinct Myeloproliferative Neoplasm Phenotype
- Author
-
Giovanni Barosi, Rita Campanelli, Margherita Massa, Paolo Catarsi, Adriana Carolei, Carlotta Abbà, Laura Villani, Umberto Magrini, Giuliana Gregato, Francesco Bertolini, Annalisa de Silvestri, Robert Peter Gale, and Vittorio Rosti
- Subjects
Hematology ,General Medicine - Abstract
Introduction: About 15% of people with a myeloproliferative neoplasm (MPN) are identified as MPN, unclassifiable using the 2016 WHO classification. Methods: We tested whether persons with platelet concentration ≥450 × 10E+9/L, bone marrow megakaryocyte morphology typical of prefibrotic/early myelofibrosis (pre-MF), and no minor criteria of pre-MF should be classified as a distinct MPN subtype, clonal megakaryocyte dysplasia with isolated thrombocytosis (CMD-IT). Results: 139 subjects meet these criteria who we compared with primary myelofibrosis (PMF) including 402 with pre-MF and 521 with overt myelofibrosis. CMD-IT subjects were more likely female and younger. They had lower frequencies of JAK2V617F compared with persons with PMF (55% vs. 70%; p < 0.001) and higher frequencies of CALR mutations (37% vs. 17%; p < 0.001). They also had lower frequency of variations associated with JAK2V617F susceptibility, JAK2 46/1 (35% vs. 47%; p = 0.021), and VEGFA rs3025039 (12% vs. 17%; p = 0.030). Subjects with CMD-IT had lower incidences of thrombotic events compared with those with pre-MF (9.7% vs. 26%; p < 0.001) and longer survival (median, not reached vs. 23 years; HR = 0.34 (0.10, 0.30); p < 0.001). Conclusion: Our data indicate CMD-IT is a distinct MPN subtype and should be included in the classification of myeloid neoplasms.
- Published
- 2022
30. Conjugated polymers mediate intracellular Ca
- Author
-
Sharon, Negri, Pawan, Faris, Gabriele, Tullii, Mauro, Vismara, Alessandro F, Pellegata, Francesco, Lodola, Gianni, Guidetti, Vittorio, Rosti, Maria Rosa, Antognazza, and Francesco, Moccia
- Subjects
Polymers ,Endothelial Cells ,TRPV Cation Channels ,Calcium ,Endoplasmic Reticulum ,Reactive Oxygen Species - Abstract
Endothelial colony forming cells (ECFCs) represent the most suitable cellular substrate to induce revascularization of ischemic tissues. Recently, optical excitation of the light-sensitive conjugated polymer, regioregular Poly (3-hexyl-thiophene), rr-P3HT, was found to stimulate ECFC proliferation and tube formation by activating the non-selective cation channel, Transient Receptor Potential Vanilloid 1 (TRPV1). Herein, we adopted a multidisciplinary approach, ranging from intracellular Ca
- Published
- 2021
31. JAK2 exon 14 skipping in patients with primary myelofibrosis: a minor splice variant modulated by the JAK2-V617F allele burden.
- Author
-
Paolo Catarsi, Vittorio Rosti, Giacomo Morreale, Valentina Poletto, Laura Villani, Roberto Bertorelli, Matteo Pedrazzini, Michele Zorzetto, Giovanni Barosi, and AGIMM investigators
- Subjects
Medicine ,Science - Abstract
Primary myelofibrosis (PMF) is an acquired clonal disease of the hematopoietic stem cell compartment, characterized by bone marrow fibrosis, anemia, splenomegaly and extramedullary hematopoiesis. About 60% of patients with PMF harbor a somatic mutation of the JAK2 gene (JAK2-V617F) in their hematopoietic lineage. Recently, a splicing isoform of JAK2, lacking exon 14 (JAK2Δ14) was described in patients affected by myeloproliferative diseases.By using a specific RT-qPCR method, we measured the ratio between the splicing isoform and the JAK2 full-length transcript (JAK2+14) in granulocytes, isolated from peripheral blood, of forty-four patients with PMF and nine healthy donors.We found that JAK2Δ14 was only slightly increased in patients and, at variance with published data, the splicing isoform was also detectable in healthy controls. We also found that, in patients bearing the JAK2-V617F mutation, the percentage of mutated alleles correlated with the observed increase in JAK2Δ14. Homozygosity for the mutation was also associated with a higher level of JAK2+14. Bioinformatic analysis indicates the possibility that the G>T transversion may interfere with the correct splicing of exon 14 by modifying a splicing regulatory sequence.Increased levels of JAK2 full-length transcript and a small but significant increase in JAK2 exon 14 skipping, are associated with the JAK2-V617F allele burden in PMF granulocytes. Our data do not confirm a previous claim that the production of the JAK2Δ14 isoform is related to the pathogenesis of PMF.
- Published
- 2015
- Full Text
- View/download PDF
32. Increased Plasma Levels of lncRNAs
- Author
-
Sebastian, Fantini, Sebastiano, Rontauroli, Stefano, Sartini, Margherita, Mirabile, Elisa, Bianchi, Filippo, Badii, Monica, Maccaferri, Paola, Guglielmelli, Tiziana, Ottone, Raffaele, Palmieri, Elena, Genovese, Chiara, Carretta, Sandra, Parenti, Selene, Mallia, Lara, Tavernari, Costanza, Salvadori, Francesca, Gesullo, Chiara, Maccari, Michela, Zizza, Alexis, Grande, Silvia, Salmoiraghi, Barbara, Mora, Leonardo, Potenza, Vittorio, Rosti, Francesco, Passamonti, Alessandro, Rambaldi, Maria Teresa, Voso, Cristina, Mecucci, Enrico, Tagliafico, Mario, Luppi, Alessandro Maria, Vannucchi, and Rossella, Manfredini
- Subjects
MPN ,lncRNAs ,biomarkers ,myelofibrosis ,prognosis ,Article - Abstract
Simple Summary Myelofibrosis (MF) displays the worst prognosis among Philadelphia-negative chronic myeloproliferative neoplasms. There is no curative therapy for MF, except for bone marrow transplantation, which however has a consistent percentage of failure. There is thus an urgent need of novel biomarkers to complement current stratification models and to enable better management of patients. To address this issue, we herein measured the plasma levels of several long noncoding RNAs (lncRNAs). Circulating lncRNAs has been already largely described as potential non-invasive biomarkers in cancers. In our study we unveiled that LINC01268, MALAT1 (both p < 0.0001) and GAS5 (p = 0.0003) plasma levels are significantly higher in MF patients if compared with healthy donors, and their increased plasma levels correlate with several detrimental features in MF. Among them, LINC01268 is an independent variable for both OS (p = 0.0297) and LFS (p = 0.0479), thus representing a putative new biomarker suitable for integrate contemporary prognostic models. Abstract Long non-coding RNAs (lncRNAs) have been recently described as key mediators in the development of hematological malignancies. In the last years, circulating lncRNAs have been proposed as a new class of non-invasive biomarkers for cancer diagnosis and prognosis and to predict treatment response. The present study is aimed to investigate the potential of circulating lncRNAs as non-invasive prognostic biomarkers in myelofibrosis (MF), the most severe among Philadelphia-negative myeloproliferative neoplasms. We detected increased levels of seven circulating lncRNAs in plasma samples of MF patients (n = 143), compared to healthy controls (n = 65). Among these, high levels of LINC01268, MALAT1 or GAS5 correlate with detrimental clinical variables, such as high count of leukocytes and CD34+ cells, severe grade of bone marrow fibrosis and presence of splenomegaly. Strikingly, high plasma levels of LINC01268 (p = 0.0018), GAS5 (p = 0.0008) or MALAT1 (p = 0.0348) are also associated with a poor overall-survival while high levels of LINC01268 correlate with a shorter leukemia-free-survival. Finally, multivariate analysis demonstrated that the plasma level of LINC01268 is an independent prognostic variable, suggesting that, if confirmed in future in an independent patients’ cohort, it could be used for further studies to design an updated classification model for MF patients.
- Published
- 2021
33. Clinical Relevance of VEGFA (rs3025039) +936 C>T Polymorphism in Primary Myelofibrosis: Susceptibility, Clinical Co-Variates, and Outcomes
- Author
-
Robert Peter Gale, Laura Villani, Adriana Carolei, Paolo Catarsi, Carlotta Abbà, Giovanni Barosi, Vittorio Rosti, Rita Campanelli, and Margherita Massa
- Subjects
VEGFA ,Male ,Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,Genotype ,Deep vein ,QH426-470 ,Gastroenterology ,Polymorphism, Single Nucleotide ,Article ,deep vein thrombosis ,chemistry.chemical_compound ,Gene Frequency ,Polymorphism (computer science) ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Genetics ,Humans ,Clinical significance ,Platelet ,Genetic Predisposition to Disease ,Myelofibrosis ,Genetics (clinical) ,Alleles ,Genetic Association Studies ,Venous Thrombosis ,vascular endothelial growth factor ,business.industry ,rs3025039 polymorphism ,Middle Aged ,medicine.disease ,Thrombosis ,Vascular endothelial growth factor ,medicine.anatomical_structure ,chemistry ,Primary Myelofibrosis ,Cytogenetic Analysis ,Mutation ,Female ,business - Abstract
We evaluated the association of VEGFA rs3025039 polymorphism with clinical co-variates and outcomes in 849 subjects with primary myelofibrosis (PMF) and 250 healthy controls. Minor T-allele frequency was higher in subjects with JAK2V617F compared with those without JAK2V617F (18% vs. 13%; p = 0.014). In subjects with JAK2V617F, the TT genotype was associated at diagnosis with lower platelet concentrations (p = 0.033), higher plasma LDH concentration (p = 0.005), higher blood CD34-positive cells (p = 0.027), lower plasma cholesterol concentration (p = 0.046), and higher concentration of high-sensitivity C-reactive protein (p = 0.018). These associations were not found in subjects with PMF without JAK2V617F. In subjects with the TT genotype, risk of death was higher compared with subjects with CC/CT genotypes (HR = 2.12 [1.03, 4.35], p = 0.041). Finally, the TT genotype was associated with higher frequency of deep vein thrombosis in typical sites (12.5% vs. 2.5%; OR = 5.46 [1.51, 19.7], p = 0.009). In conclusion, in subjects with PMF, the VEGFA rs3025039 CT or TT genotypes are more common in those with JAK2V617F than in those without JAK2V67F mutation and are associated with disease severity, poor prognosis, and risk of deep vein thrombosis.
- Published
- 2021
34. A prospective evaluation of pegylated interferon alfa-2a therapy in patients with polycythemia vera and essential thrombocythemia with a prior splanchnic vein thrombosis
- Author
-
John Mascarenhas, Marina Kremyanskaya, Mohamed E. Salama, Abdulraheem Yacoub, Craig M. Kessler, Dmitriy Berenzon, Ellen K. Ritchie, Joseph Vadakara, Damiano Rondelli, Rona Singer Weinberg, Elliot F. Winton, Richard T. Silver, Alessandro M. Vannucchi, Raajit K. Rampal, Ruben A. Mesa, Vesna Najfeld, Alessandro Rambaldi, Maria Chiara Finazzi, Joseph Tripodi, Robert S. Hoffman, Elizabeth O. Hexner, David S. Leibowitz, Murat O. Arcasoy, J. T. Prchal, Vittorio Rosti, Noushin Farnoud, Amylou C. Dueck, Rosalind Catchatourian, Maria R. Baer, Judith D. Goldberg, Lonette Sandy, and Heidi E. Kosiorek
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Gastroenterology ,Article ,Polyethylene Glycols ,03 medical and health sciences ,Mesenteric Veins ,0302 clinical medicine ,Polycythemia vera ,Pegylated interferon ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Prospective Studies ,Vein ,Prospective cohort study ,Myelofibrosis ,Polycythemia Vera ,Venous Thrombosis ,Acute leukemia ,Essential thrombocythemia ,business.industry ,Interferon-alpha ,Hematology ,medicine.disease ,Recombinant Proteins ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Splanchnic vein thrombosis ,030220 oncology & carcinogenesis ,business ,Thrombocythemia, Essential ,medicine.drug - Abstract
Essential thrombocythemia (ET) and polycythemia vera (PV) are myeloproliferative neoplasms (MPNs) characterized by an increased risk of developing venous and arterial thromboses and of evolution to myelofibrosis or acute leukemia (1). MPNs are recognized as a major cause of splanchnic vein thromboses (SVT), which include hepatic, splenic, portal and mesenteric vein thromboses (2). Patients with SVT are at an increased risk of developing bleeding events and arterial thrombotic events (3), as well as recurrent SVT and the use of anticoagulation and cytoreduction do not clearly modify this risk (3–5). Management of SVT in MPN patients remains a challenge and most therapeutic recommendations are based on retrospective analyses (6). Recently, De Stefano reported that hydroxyurea failed to prevent recurrent thromboses in the splanchnic vasculature in patients with a prior SVT (7). We conducted a prospective, open-label, multi-center phase II clinical trial of Pegylated Interferon Alfa-2a (PEG) in 20 subjects with SVT and an MPN, who represented a cohort of patients who participated in the Myeloproliferative Disorders - Research Consortium (MPD-RC) 111 trial (). MPD-RC 111 was a prospective study of patients with high-risk ET/PV who were resistant or intolerant to hydroxyurea (HU), but prior HU therapy was not a requirement for patients to enter the SVT cohort. Here we report the outcomes of the 20 SVT patients who were treated with PEG.
- Published
- 2019
- Full Text
- View/download PDF
35. Enhanced expression of Stim, Orai, and TRPC transcripts and proteins in endothelial progenitor cells isolated from patients with primary myelofibrosis.
- Author
-
Silvia Dragoni, Umberto Laforenza, Elisa Bonetti, Marta Reforgiato, Valentina Poletto, Francesco Lodola, Cinzia Bottino, Daniele Guido, Alessandra Rappa, Sumedha Pareek, Mario Tomasello, Maria Rosa Guarrera, Maria Pia Cinelli, Adele Aronica, Germano Guerra, Giovanni Barosi, Franco Tanzi, Vittorio Rosti, and Francesco Moccia
- Subjects
Medicine ,Science - Abstract
BackgroundAn increase in the frequency of circulating endothelial colony forming cells (ECFCs), the only subset of endothelial progenitor cells (EPCs) truly belonging to the endothelial phenotype, occurs in patients affected by primary myelofibrosis (PMF). Herein, they might contribute to the enhanced neovascularisation of fibrotic bone marrow and spleen. Store-operated Ca2+ entry (SOCE) activated by the depletion of the inositol-1,4,5-trisphosphate (InsP3)-sensitive Ca2+ store drives proliferation in ECFCs isolated from both healthy donors (N-ECFCs) and subjects suffering from renal cellular carcinoma (RCC-ECFCs). SOCE is up-regulated in RCC-ECFCs due to the over-expression of its underlying molecular components, namely Stim1, Orai1, and TRPC1.Methodology/principal findingsWe utilized Ca2+ imaging, real-time polymerase chain reaction, western blot analysis and functional assays to evaluate molecular structure and the functional role of SOCE in ECFCs derived from PMF patients (PMF-ECFCs). SOCE, induced by either pharmacological (i.e. cyclopiazonic acid or CPA) or physiological (i.e. ATP) stimulation, was significantly higher in PMF-ECFCs. ATP-induced SOCE was inhibited upon blockade of the phospholipase C/InsP3 signalling pathway with U73111 and 2-APB. The higher amplitude of SOCE was associated to the over-expression of the transcripts encoding for Stim2, Orai2-3, and TRPC1. Conversely, immunoblotting revealed that Stim2 levels remained constant as compared to N-ECFCs, while Stim1, Orai1, Orai3, TRPC1 and TRPC4 proteins were over-expressed in PMF-ECFCs. ATP-induced SOCE was inhibited by BTP-2 and low micromolar La3+ and Gd3+, while CPA-elicited SOCE was insensitive to Gd3+. Finally, BTP-2 and La3+ weakly blocked PMF-ECFC proliferation, while Gd3+ was ineffective.ConclusionsTwo distinct signalling pathways mediate SOCE in PMF-ECFCs; one is activated by passive store depletion and is Gd3+-resistant, while the other one is regulated by the InsP3-sensitive Ca2+ pool and is inhibited by Gd3+. Unlike N- and RCC-ECFCs, the InsP3-dependent SOCE does not drive PMF-ECFC proliferation.
- Published
- 2014
- Full Text
- View/download PDF
36. A subpopulation of circulating endothelial cells express CD109 and is enriched in the blood of cancer patients.
- Author
-
Patrizia Mancuso, Angelica Calleri, Giuliana Gregato, Valentina Labanca, Jessica Quarna, Pierluigi Antoniotti, Lucia Cuppini, Gaetano Finocchiaro, Marica Eoli, Vittorio Rosti, and Francesco Bertolini
- Subjects
Medicine ,Science - Abstract
BACKGROUND:The endothelium is not a homogeneous organ. Endothelial cell heterogeneity has been described at the level of cell morphology, function, gene expression, and antigen composition. As a consequence of the genetic, transcriptome and surrounding environment diversity, endothelial cells from different vascular beds have differentiated functions and phenotype. Detection of circulating endothelial cells (CECs) by flow cytometry is an approach widely used in cancer patients, and their number, viability and kinetic is a promising tool to stratify patient receiving anti-angiogenic treatment. METHODOLOGY/PRINCIPAL FINDINGS:Currently CECs are identified as positive for a nuclear binding antigen (DNA+), negative for the pan leukocyte marker CD45, and positive for CD31 and CD146. Following an approach recently validated in our laboratory, we investigated the expression of CD109 on CECs from the peripheral blood of healthy subject and cancer patients. The endothelial nature of these cells was validated by RT-PCR for the presence of m-RNA level of CDH5 (Ve-Cadherin) and CLDN5 (Claudin5), two endothelial specific transcripts. Before treatment, significantly higher levels of CD109+ CECs and viable CD109+CECs were found in breast cancer patients and glioblastoma patients compared to healthy controls, and their number significantly decreased after treatment. Higher levels of endothelial specific transcripts expressed in developing endothelial cells CLEC14a, TMEM204, ARHGEF15, GPR116, were observed in sorted CD109+CECs when compared to sorted CD146+CECs, suggesting that these genes can play an important role not only during embryogenesis but also in adult angiogenesis. Interestingly, mRNA levels of TEM8 (identified as Antrax Toxin Receptor1, Antrax1) were expressed in CD109+CECs+ but not in CD146+CECs. CONCLUSION:Taken together our results suggest that CD109 represent a rare population of circulating tumor endothelial cells, that play a potentially useful prognostic role in patients with glioblastoma. The role of CD109 expression in cancer vessel-specific endothelial cells deserves to be further investigated by gene expression studies.
- Published
- 2014
- Full Text
- View/download PDF
37. Systemic lupus erythematosus, endothelial progenitor cells and intracellular Ca 2+ signaling A novel approach for an old disease
- Author
-
Francesco Moccia, Ricard Cervera, Germano Guerra, Vittorio Rosti, Claudia Mendoza-Pinto, Mario García-Carrasco, Roberto Berra-Romani, Klara Komici, Sharon Negri, and Pawan Faris
- Subjects
Myeloid ,Angiogenesis ,Immunology ,Pathogenesis ,03 medical and health sciences ,Paracrine signalling ,Systemic lupus erythematosus ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Endothelial dysfunction ,Progenitor cell ,Endothelial progenitor cells ,Intracellular Ca(2+) signaling ,030304 developmental biology ,Tube formation ,0303 health sciences ,business.industry ,medicine.disease ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,business ,Homeostasis - Abstract
Systemic lupus erythematosus (SLE) is an autoimmune multisystem disease featured by an increased cardiovascular risk that may lead to premature patient's death. It has been demonstrated that SLE patients suffer from early onset endothelial dysfunction which is due to the impairment of endogenous vascular repair mechanisms. Vascular integrity and homeostasis are maintained by endothelial progenitor cells (EPCs), which are mobilized in response to endothelial injury to replace damaged endothelial cells. Two main EPCs subpopulations exist in peripheral blood: endothelial colony forming cells (ECFCs), which represent truly endothelial precursors and can physically engraft within neovessels, and myeloid angiogenic cells (MACs), which sustain angiogenesis in a paracrine manner. Emerging evidence indicates that ECFCs/MACs are down-regulated and display compromised angiogenic activity in SLE, thereby contributing to the pathogenesis of this disease. Intracellular calcium (Ca2+) signaling plays a crucial role in maintaining vascular integrity by stimulating migration, proliferation and tube formation in both ECFCs and MACs. Herein, we illustrate the evidences that support the role played by EPCs dysfunction in SLE. Subsequently, we discuss about the hypothesis that the Ca2+ handling machinery is compromised in SLE-derived ECFCs and MACs, thereby resulting in their reduced pro-angiogenic activity. Finally, we speculate about the proposal to exploit intracellular Ca2+ signaling to improve ECFCs' reparative phenotype and suggest this strategy as a new approach to treat SLE patients.
- Published
- 2021
38. Impact of the rs1024611 Polymorphism of CCL2 on the Pathophysiology and Outcome of Primary Myelofibrosis
- Author
-
Antonio Percesepe, Giovanni Roti, Margherita Massa, Marco Vitale, Sabrina Bonomini, Giovanni Barosi, Elena Masselli, Laura Villani, Giuliana Gobbi, Giulia Pozzi, Cecilia Carubbi, Vittorio Rosti, and Rita Campanelli
- Subjects
0301 basic medicine ,Cancer Research ,CCR2 ,Chemokine ,Ruxolitinib ,medicine.medical_treatment ,Single-nucleotide polymorphism ,Article ,myeloproliferative neoplasms ,03 medical and health sciences ,single nucleotide polymorphisms ,0302 clinical medicine ,Polymorphism (computer science) ,Genotype ,medicine ,SNP ,RC254-282 ,biology ,Akt ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Cytokine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,primary myelofibrosis ,CCL2 ,medicine.drug - Abstract
Single nucleotide polymorphisms (SNPs) can modify the individual pro-inflammatory background and may therefore have relevant implications in the MPN setting, typified by aberrant cytokine production. In a cohort of 773 primary myelofibrosis (PMF), we determined the contribution of the rs1024611 SNP of CCL2—one of the most potent immunomodulatory chemokines—to the clinical and biological characteristics of the disease, demonstrating that male subjects carrying the homozygous genotype G/G had an increased risk of PMF and that, among PMF patients, the G/G genotype is an independent prognostic factor for reduced overall survival. Functional characterization of the SNP and the CCL2-CCR2 axis in PMF showed that (i) homozygous PMF cells are the highest chemokine producers as compared to the other genotypes, (ii) PMF CD34+ cells are a selective target of CCL2, since they uniquely express CCR2 (CCL2 receptor), (iii) activation of the CCL2-CCR2 axis boosts pro-survival signals induced by driver mutations via Akt phosphorylation, (iv) ruxolitinib effectively counteracts CCL2 production and down-regulates CCR2 expression in PMF cells. In conclusion, the identification of the role of the CCL2/CCR2 chemokine system in PMF adds a novel element to the pathophysiological picture of the disease, with clinical and therapeutic implications.
- Published
- 2021
39. Nicotinic acid adenine dinucleotide phosphate activates two‐pore channel TPC1 to mediate lysosomal Ca 2+ release in endothelial colony‐forming cells
- Author
-
Germano Guerra, Antonio De Luca, Luigi Ambrosone, Pawan Faris, Francesca Di Nezza, Sharon Negri, Giorgia Pellavio, Francesco Moccia, Estella Zuccolo, Vittorio Rosti, Umberto Laforenza, Moccia, F., Zuccolo, E., Di Nezza, F., Pellavio, G., Faris, P. S., Negri, S., De Luca, A., Laforenza, U., Ambrosone, L., Rosti, V., and Guerra, G.
- Subjects
0301 basic medicine ,Ca ,Nigericin ,Physiology ,Clinical Biochemistry ,NAADP ,2+ ,two-pore channel 1 ,Nitric oxide ,endothelial colony-forming cells ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Ca2+ signaling ,VEGF ,Receptor ,Electrochemical gradient ,Nicotinic acid adenine dinucleotide phosphate ,Endoplasmic reticulum ,Cell Biology ,Cell biology ,030104 developmental biology ,Two-pore channel ,chemistry ,030220 oncology & carcinogenesis ,endothelial colony-forming cell ,signaling ,Intracellular - Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is the most recently discovered Ca2+ -releasing messenger that increases the intracellular Ca2+ concentration by mobilizing the lysosomal Ca2+ store through two-pore channels 1 (TPC1) and 2 (TPC2). NAADP-induced lysosomal Ca2+ release regulates multiple endothelial functions, including nitric oxide release and proliferation. A sizeable acidic Ca2+ pool endowed with TPC1 is also present in human endothelial colony-forming cells (ECFCs), which represent the only known truly endothelial precursors. Herein, we sought to explore the role of the lysosomal Ca2+ store and TPC1 in circulating ECFCs by harnessing Ca2+ imaging and molecular biology techniques. The lysosomotropic agent, Gly-Phe β-naphthylamide, and nigericin, which dissipates the proton gradient which drives Ca2+ sequestration by acidic organelles, caused endogenous Ca2+ release in the presence of a replete inositol-1,4,5-trisphosphate (InsP3 )-sensitive endoplasmic reticulum (ER) Ca2+ pool. Likewise, the amount of ER releasable Ca2+ was reduced by disrupting lysosomal Ca2+ content. Liposomal delivery of NAADP induced a transient Ca2+ signal that was abolished by disrupting the lysosomal Ca2+ store and by pharmacological and genetic blockade of TPC1. Pharmacological manipulation revealed that NAADP-induced Ca2+ release also required ER-embedded InsP3 receptors. Finally, NAADP-induced lysosomal Ca2+ release was found to trigger vascular endothelial growth factor-induced intracellular Ca2+ oscillations and proliferation, while it did not contribute to adenosine-5'-trisphosphate-induced Ca2+ signaling. These findings demonstrated that NAADP-induced TPC1-mediated Ca2+ release can selectively be recruited to induce the Ca2+ response to specific cues in circulating ECFCs.
- Published
- 2021
- Full Text
- View/download PDF
40. Conjugated polymers mediate intracellular Ca2+ signals in circulating endothelial colony forming cells through the reactive oxygen species-dependent activation of Transient Receptor Potential Vanilloid 1 (TRPV1)
- Author
-
Pawan Faris, Francesco Moccia, Mauro Vismara, Alessandro F. Pellegata, Gianni Francesco Guidetti, Sharon Negri, Vittorio Rosti, Francesco Lodola, Gabriele Tullii, Maria Rosa Antognazza, Negri, S, Faris, P, Tullii, G, Vismara, M, Pellegata, A, Lodola, F, Guidetti, G, Rosti, V, Antognazza, M, and Moccia, F
- Subjects
Physiology ,Polymers ,TRPV1 ,Endothelial colony forming cells ,TRPV Cation Channels ,Conjugated polymers ,Endoplasmic Reticulum ,Endothelial colony forming cell ,Cell membrane ,03 medical and health sciences ,Transient receptor potential channel ,Optical stimulation ,0302 clinical medicine ,Extracellular ,medicine ,Receptor ,Molecular Biology ,030304 developmental biology ,Tube formation ,0303 health sciences ,Chemistry ,Endoplasmic reticulum ,Conjugated polymer ,Endothelial Cells ,Cell Biology ,Transient receptor potential vanilloid 1 ,medicine.anatomical_structure ,Biophysics ,Reactive oxygen specie ,Calcium ,Reactive Oxygen Species ,030217 neurology & neurosurgery ,Intracellular - Abstract
Endothelial colony forming cells (ECFCs) represent the most suitable cellular substrate to induce revascularization of ischemic tissues. Recently, optical excitation of the light-sensitive conjugated polymer, regioregular Poly (3-hexyl-thiophene), rr-P3HT, was found to stimulate ECFC proliferation and tube formation by activating the non-selective cation channel, Transient Receptor Potential Vanilloid 1 (TRPV1). Herein, we adopted a multidisciplinary approach, ranging from intracellular Ca2+ imaging to pharmacological manipulation and genetic suppression of TRPV1 expression, to investigate the effects of photoexcitation on intracellular Ca2+ concentration ([Ca2+]i) in circulating ECFCs plated on rr-P3HT thin films. Polymer-mediated optical excitation induced a long-lasting increase in [Ca2+]i that could display an oscillatory pattern at shorter light stimuli. Pharmacological and genetic manipulation revealed that the Ca2+ response to light was triggered by extracellular Ca2+ entry through TRPV1, whose activation required the production of reactive oxygen species at the interface between rr-P3HT and the cell membrane. Light-induced TRPV1-mediated Ca2+ entry was able to evoke intracellular Ca2+ release from the endoplasmic reticulum through inositol-1,4,5-trisphosphate receptors, followed by store-operated Ca2+ entry on the plasma membrane. These data show that TRPV1 may serve as a decoder at the interface between rr-P3HT thin films and ECFCs to translate optical excitation in pro-angiogenic Ca2+ signals.
- Published
- 2021
- Full Text
- View/download PDF
41. Long-term safety and efficacy of givinostat in polycythemia vera: 4-year mean follow up of three phase 1/2 studies and a compassionate use program
- Author
-
Bruno Martino, Sara Manzoni, Vincenzo Martinelli, Alessandra Iurlo, Attilio Guarini, Mary Frances McMullin, Marianna De Muro, Silvia Di Tollo, Alessandro M. Vannucchi, Marco Ruggeri, Axel Nogai, Vittorio Rosti, Paolo Bettica, Alessandro Rambaldi, Stefania Luciani, Nikolas von Bubnoff, Alessandra Ricco, Rambaldi, A., Iurlo, A., Vannucchi, A. M., Martino, B., Guarini, A., Ruggeri, M., von Bubnoff, N., De Muro, M., Mcmullin, M. F., Luciani, S., Martinelli, V., Nogai, A., Rosti, V., Ricco, A., Bettica, P., Manzoni, S., and Di Tollo, S.
- Subjects
Adult ,Male ,medicine.medical_specialty ,Population ,Phases of clinical research ,Drug development ,Gene mutation ,lcsh:RC254-282 ,Article ,Efficacy ,chemistry.chemical_compound ,Myeloproliferative disease ,Polycythemia vera ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Givinostat ,education ,Polycythemia Vera ,Myeloproliferative neoplasm ,Aged ,education.field_of_study ,business.industry ,Hematology ,Middle Aged ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Histone Deacetylase Inhibitors ,Treatment Outcome ,Oncology ,Tolerability ,chemistry ,Female ,Carbamates ,business ,Follow-Up Studies - Abstract
Polycythemia vera (PV) is a BCR-ABL1-negative myeloproliferative neoplasm (MPN) characterized by excessive proliferation of erythroid, myeloid, and megakaryocytic components in the bone marrow, mainly due to a Janus kinase 2 gene mutation (JAK2V617F). Givinostat, a histone-deacetylase inhibitor that selectively targets JAK2V617F cell growth, has demonstrated good efficacy and safety in three phase 1/2 studies in patients with PV. This manuscript focuses on the 4-year mean (2.8 year median) follow-up of an open-label, long-term study that enrolled 51 patients with PV (out of a total of 54 with MPN) who received clinical benefit from givinostat in these previous studies or on compassionate use, and who continued to receive givinostat at the last effective and tolerated dose. The primary objectives are to determine givinostat’s long-term safety and tolerability, and efficacy evaluated by the investigators according to internationally recognized response criteria. During follow-up, only 10% of PV patients reported Grade 3 treatment-related adverse events (AEs), while none had Grade 4 or 5 treatment-related AEs. The overall response rate for the duration of follow-up was always greater than 80% in patients with PV. In conclusion, givinostat demonstrated a good safety and efficacy profile in patients with PV, data supporting long-term use in this population.
- Published
- 2021
- Full Text
- View/download PDF
42. No association between the XPD Lys751Gln (rs13181) polymorphism and disease phenotype or leukemic transformation in primary myelofibrosis
- Author
-
Valentina Poletto, Laura Villani, Paolo Catarsi, Rita Campanelli, Margherita Massa, Alessandro M. Vannucchi, Vittorio Rosti, and Giovanni Barosi
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2013
- Full Text
- View/download PDF
43. Involvement of TGFβ1 in autocrine regulation of proplatelet formation in healthy subjects and patients with primary myelofibrosis
- Author
-
Stefania Badalucco, Christian Andrea Di Buduo, Rita Campanelli, Isabella Pallotta, Paolo Catarsi, Vittorio Rosti, David L. Kaplan, Giovanni Barosi, Margherita Massa, and Alessandra Balduini
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Megakaryocytes release platelets into the bloodstream by elongating proplatelets. In this study, we showed that human megakaryocytes constitutively release Transforming Growth Factor β1 and express its receptors. Importantly, Transforming Growth Factor β1 downstream signaling, through SMAD2/3 phosphorylation, was shown to be active in megakaryocytes extending proplatelets, indicating a type of autocrine stimulation on megakaryocyte development. Furthermore, inactivation of Transforming Growth Factor β1 signaling, by the receptor inhibitors SB431542 and Stemolecule ALK5 inhibitor, determined a significant decrease in proplatelet formation. Recent studies indicated a crucial role of Transforming Growth Factor β1 in the pathogenesis of primary myelofibrosis. We demonstrated that primary myelofibrosis-derived megakaryocytes expressed increased levels of bioactive Transforming Growth Factor β1; however, higher levels of released Transforming Growth Factor β1 did not lead to enhanced activation of downstream pathways. Overall, these data propose Transforming Growth Factor β1 as a new element in the autocrine regulation of proplatelet formation in vitro. Despite the increase in Transforming Growth Factor β1 this mechanism seems to be preserved in primary myelofibrosis.
- Published
- 2013
- Full Text
- View/download PDF
44. JAK2 V617F genotype is a strong determinant of blast transformation in primary myelofibrosis.
- Author
-
Giovanni Barosi, Valentina Poletto, Margherita Massa, Rita Campanelli, Laura Villani, Elisa Bonetti, Gianluca Viarengo, Paolo Catarsi, Catherine Klersy, and Vittorio Rosti
- Subjects
Medicine ,Science - Abstract
PurposeThe influence of JAK2 V617F mutation on blast transformation (BT) and overall survival (OS) in primary myelofibrosis (PMF) is controversial. In a large cohort of patients we applied competing risks analysis for studying the influence of JAK2V617F mutation on BT in PMF.Patients and methodsIn 462 PMF-fibrotic type patients (bone marrow [BM] fibrosis grade >0) we computed the incidence of BT and death in the framework of Cox regression analysis and of Fine and Gray competing risks analysis for BT.ResultsAt the Cox regression analysis, having either a wild-type (wt) or a homozygous JAK2V617F genotype were factors for BT (HR, 1.98 and 2.04, respectively, with respect to the heterozygous genotype), but not for OS. At the competing risks regression analysis, the risk for BT in wt and homozygous V617F patients increased with respect to Cox analysis, giving a sHR of 2.17 and 2.12, respectively. Correcting the results for the variables that could have influence on BT, JAK2V617F wt and homozygous genotypes remained independently associated with BT. In a validation cohort of 133 independent cases with PMF-prefibrotic type (BM fibrosis grade = 0), the BT predictive model including JAK2V617F genotype and older age retained high discriminant capacity (C statistics, 0.70; 95% CI, 0.47 to 0.92).ConclusionThe accumulation of mutated alleles in the JAK2V617F clone or the selective acquisition of a proliferative advantage in the wt clone are two relevant routes to BT in PMF. The influence of these results on treatment decisions with anti-JAK2 agents should be tested.
- Published
- 2013
- Full Text
- View/download PDF
45. Biological, functional and genetic characterization of bone marrow-derived mesenchymal stromal cells from pediatric patients affected by acute lymphoblastic leukemia.
- Author
-
Antonella Conforti, Simone Biagini, Francesca Del Bufalo, Pietro Sirleto, Adriano Angioni, Nadia Starc, Giuseppina Li Pira, Francesca Moretta, Alessandra Proia, Benedetta Contoli, Silvia Genovese, Claudia Ciardi, Maria Antonietta Avanzini, Vittorio Rosti, Francesco Lo-Coco, Franco Locatelli, and Maria Ester Bernardo
- Subjects
Medicine ,Science - Abstract
Alterations in hematopoietic microenvironment of acute lymphoblastic leukemia patients have been claimed to occur, but little is known about the components of marrow stroma in these patients. In this study, we characterized mesenchymal stromal cells (MSCs) isolated from bone marrow (BM) of 45 pediatric patients with acute lymphoblastic leukemia (ALL-MSCs) at diagnosis (day+0) and during chemotherapy treatment (days: +15; +33; +78), the time points being chosen according to the schedule of BM aspirates required by the AIEOP-BFM ALL 2009 treatment protocol. Morphology, proliferative capacity, immunophenotype, differentiation potential, immunomodulatory properties and ability to support long-term hematopoiesis of ALL-MSCs were analysed and compared with those from 41 healthy donors (HD-MSCs). ALL-MSCs were also genetically characterized through array-CGH, conventional karyotyping and FISH analysis. Moreover, we compared ALL-MSCs generated at day+0 with those isolated during chemotherapy. Morphology, immunophenotype, differentiation potential and in vitro life-span did not differ between ALL-MSCs and HD-MSCs. ALL-MSCs showed significantly lower proliferative capacity (p
- Published
- 2013
- Full Text
- View/download PDF
46. Therapeutic Potential of Endothelial Colony-Forming Cells in Ischemic Disease: Strategies to Improve their Regenerative Efficacy
- Author
-
Francesco Moccia, Vittorio Rosti, Pawan Faris, Angelica Perna, Sharon Negri, and Germano Guerra
- Subjects
0301 basic medicine ,Angiogenesis ,Cell- and Tissue-Based Therapy ,Review ,Disease ,030204 cardiovascular system & hematology ,Bioinformatics ,Neovascularization ,lcsh:Chemistry ,0302 clinical medicine ,Ischemia ,cardiovascular disease ,therapeutic angiogenesis ,Medicine ,genetic modification ,Myocardial infarction ,lcsh:QH301-705.5 ,Spectroscopy ,Endothelial Progenitor Cells ,endothelial colony forming cells ,Chemotaxis ,Disease Management ,Cell Differentiation ,General Medicine ,pharmacological conditioning ,3. Good health ,Computer Science Applications ,Phenotype ,medicine.anatomical_structure ,ischemic disorders ,Disease Susceptibility ,medicine.symptom ,Signal Transduction ,Blood vessel ,Neovascularization, Physiologic ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,Vasculogenesis ,Animals ,Humans ,Therapeutic angiogenesis ,Physical and Theoretical Chemistry ,Molecular Biology ,Protein kinase B ,business.industry ,Organic Chemistry ,Mesenchymal Stem Cells ,medicine.disease ,signaling pathways ,030104 developmental biology ,Gene Expression Regulation ,lcsh:Biology (General) ,lcsh:QD1-999 ,business ,Biomarkers ,Stem Cell Transplantation - Abstract
Cardiovascular disease (CVD) comprises a range of major clinical cardiac and circulatory diseases, which produce immense health and economic burdens worldwide. Currently, vascular regenerative surgery represents the most employed therapeutic option to treat ischemic disorders, even though not all the patients are amenable to surgical revascularization. Therefore, more efficient therapeutic approaches are urgently required to promote neovascularization. Therapeutic angiogenesis represents an emerging strategy that aims at reconstructing the damaged vascular network by stimulating local angiogenesis and/or promoting de novo blood vessel formation according to a process known as vasculogenesis. In turn, circulating endothelial colony-forming cells (ECFCs) represent truly endothelial precursors, which display high clonogenic potential and have the documented ability to originate de novo blood vessels in vivo. Therefore, ECFCs are regarded as the most promising cellular candidate to promote therapeutic angiogenesis in patients suffering from CVD. The current briefly summarizes the available information about the origin and characterization of ECFCs and then widely illustrates the preclinical studies that assessed their regenerative efficacy in a variety of ischemic disorders, including acute myocardial infarction, peripheral artery disease, ischemic brain disease, and retinopathy. Then, we describe the most common pharmacological, genetic, and epigenetic strategies employed to enhance the vasoreparative potential of autologous ECFCs by manipulating crucial pro-angiogenic signaling pathways, e.g., extracellular-signal regulated kinase/Akt, phosphoinositide 3-kinase, and Ca2+ signaling. We conclude by discussing the possibility of targeting circulating ECFCs to rescue their dysfunctional phenotype and promote neovascularization in the presence of CVD.
- Published
- 2020
47. Therapeutic Potential of Endothelial Colony Forming Cells in Ischemic Disease: Strategies to Improve Their Regenerative Efficacy
- Author
-
Angelica Perna, Pawan Faris, Germano Guerra, Francesco Moccia, Sharon Negri, and Vittorio Rosti
- Subjects
0303 health sciences ,Angiogenesis ,business.industry ,Disease ,030204 cardiovascular system & hematology ,Bioinformatics ,medicine.disease ,3. Good health ,Neovascularization ,03 medical and health sciences ,cell_developmental_biology ,0302 clinical medicine ,Vasculogenesis ,medicine.anatomical_structure ,medicine ,Therapeutic angiogenesis ,Myocardial infarction ,medicine.symptom ,business ,Protein kinase B ,030304 developmental biology ,Blood vessel - Abstract
Cardiovascular disease (CVD) comprises a range of major clinical cardiac and circulatory diseases, which produce immense health and economic burdens worldwide. Currently, vascular regenerative surgery represents the most employed therapeutic option to treat ischemic disorders, even though not all the patients are amenable to surgical revascularization. Therefore, more efficient therapeutic approaches are urgently required to promote neovascularization. Therapeutic angiogenesis represents an emerging strategy that aims at reconstructing the damaged vascular network by stimulating local angiogenesis and/or promoting de novo blood vessel formation according to a process known as vasculogenesis. In turn, circulating endothelial colony-forming cells (ECFCs) represent truly endothelial precursors, which display high clonogenic potential and have the documented ability to originate de novo blood vessels in vivo. Therefore, ECFCs are regarded as the most promising cellular candidate to promote therapeutic angiogenesis in patients suffering from CVD. The current briefly summarizes the available information about the origin and characterization of ECFCs and then widely illustrates the preclinical studies that assessed their regenerative efficacy in a variety of ischemic disorders, including acute myocardial infarction, peripheral artery disease, ischemic brain disease, and retinopathy. Then, we describe the most common pharmacological, genetic, and epigenetic strategies employed to enhance the vasoreparative potential of autologous ECFCs by manipulating crucial pro-angiogenic signaling pathways, e.g., extracellular-signal regulated kinase/Akt, phosphoinositide 3-kinase, and Ca2+ signaling. We conclude by discussing the possibility of targeting circulating ECFCs to rescue their dysfunctional phenotype and promote neovascularization in the presence of CVD.
- Published
- 2020
48. Increased B4GALT1 expression is associated with platelet surface galactosylation and thrombopoietin plasma levels in MPNs
- Author
-
Vittorio Rosti, Vittorio Abbonante, Karin M. Hoffmeister, Alessandra Balduini, Christian A. Di Buduo, and Silvia Giannini
- Subjects
Blood Platelets ,Myeloproliferative Disorders ,Myeloid Neoplasia ,Janus kinase 1 ,Immunology ,Galactose ,Cell Biology ,Hematology ,Biology ,Galactosyltransferases ,Biochemistry ,Up-Regulation ,Haematopoiesis ,Thrombopoietin ,Mutation ,Cancer research ,Humans ,Platelet ,Thrombopoiesis ,Stem cell ,Janus kinase ,Megakaryocytes ,Megakaryopoiesis - Abstract
Aberrant megakaryopoiesis is a hallmark of the myeloproliferative neoplasms (MPNs), a group of clonal hematological malignancies originating from hematopoietic stem cells, leading to an increase in mature blood cells in the peripheral blood. Sialylated derivatives of the glycan structure β4-N-acetyllactosamine (Galβ1,4GlcNAc or type-2 LacNAc, hereafter referred to as LacNAc) regulate platelet life span, hepatic thrombopoietin (TPO) production, and thrombopoiesis. We found increased TPO plasma levels in MPNs with high allele burden of the mutated clones. Remarkably, platelets isolated from MPNs had a significant increase in LacNAc expression that correlated with the high allele burden regardless of the underlying identified mutation. Megakaryocytes derived in vitro from these patients showed an increased expression of the B4GALT1 gene encoding β-1,4-galactosyltransferase 1 (β4GalT1). Consistently, megakaryocytes from MPN showed increased LacNAc expression relative to healthy controls, which was counteracted by the treatment with a Janus kinase 1/2 inhibitor. Altered expression of B4GALT1 in mutant megakaryocytes can lead to the production of platelets with aberrant galactosylation, which in turn promote hepatic TPO synthesis regardless of platelet mass. Our findings provide a new paradigm for understanding aberrant megakaryopoiesis in MPNs and identify β4GalT1 as a potential actionable target for therapy.
- Published
- 2020
49. Nicotinic acid adenine dinucleotide phosphate activates two-pore channel TPC1 to mediate lysosomal Ca
- Author
-
Francesco, Moccia, Estella, Zuccolo, Francesca, Di Nezza, Giorgia, Pellavio, Pawan S, Faris, Sharon, Negri, Antonio, De Luca, Umberto, Laforenza, Luigi, Ambrosone, Vittorio, Rosti, and Germano, Guerra
- Subjects
Vascular Endothelial Growth Factor A ,Endothelial Cells ,Humans ,Calcium ,Calcium Channels ,Calcium Signaling ,Endoplasmic Reticulum ,Lysosomes ,NADP ,Cell Line - Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is the most recently discovered Ca
- Published
- 2020
50. Endothelial TRPV1 as an Emerging Molecular Target to Promote Therapeutic Angiogenesis
- Author
-
Maria Rosa Antognazza, Vittorio Rosti, Sharon Negri, Francesco Moccia, Francesco Lodola, Pawan Faris, Negri, S, Faris, P, Rosti, V, Antognazza, M, Lodola, F, and Moccia, F
- Subjects
endothelial colony forming cell ,Angiogenesis ,vascular endothelial cells ,TRPV1 ,Neovascularization, Physiologic ,TRPV Cation Channels ,Review ,Models, Biological ,03 medical and health sciences ,Transient receptor potential channel ,photostimulation ,0302 clinical medicine ,Vasculogenesis ,Ca2+ signaling ,therapeutic angiogenesis ,therapeutic angiogenesi ,medicine ,Animals ,Humans ,simvastatin ,Therapeutic angiogenesis ,Molecular Targeted Therapy ,organic semiconductors ,lcsh:QH301-705.5 ,030304 developmental biology ,endothelial colony forming cells ,0303 health sciences ,Chemistry ,organic semiconductor ,musculoskeletal, neural, and ocular physiology ,Endothelial Cells ,General Medicine ,Cell biology ,Endothelial stem cell ,medicine.anatomical_structure ,evodiamine ,lcsh:Biology (General) ,nervous system ,vascular endothelial cell ,erythropoietin ,030217 neurology & neurosurgery ,Intracellular ,Blood vessel ,Signal Transduction - Abstract
Therapeutic angiogenesis represents an emerging strategy to treat ischemic diseases by stimulating blood vessel growth to rescue local blood perfusion. Therefore, injured microvasculature may be repaired by stimulating resident endothelial cells or circulating endothelial colony forming cells (ECFCs) or by autologous cell-based therapy. Endothelial Ca2+signals represent a crucial player in angiogenesis and vasculogenesis; indeed, several angiogenic stimuli induce neovessel formation through an increase in intracellular Ca2+concentration. Several members of the Transient Receptor Potential (TRP) channel superfamily are expressed and mediate Ca2+-dependent functions in vascular endothelial cells and in ECFCs, the only known truly endothelial precursor. TRP Vanilloid 1 (TRPV1), a polymodal cation channel, is emerging as an important player in endothelial cell migration, proliferation, and tubulogenesis, through the integration of several chemical stimuli. Herein, we first summarize TRPV1 structure and gating mechanisms. Next, we illustrate the physiological roles of TRPV1 in vascular endothelium, focusing our attention on how endothelial TRPV1 promotes angiogenesis. In particular, we describe a recent strategy to stimulate TRPV1-mediated pro-angiogenic activity in ECFCs, in the presence of a photosensitive conjugated polymer. Taken together, these observations suggest that TRPV1 represents a useful target in the treatment of ischemic diseases.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.